EP1926813B2 - Zellpopulationen mit immunregulationsaktivität, verfahren zur isolierung und verwendungen - Google Patents

Zellpopulationen mit immunregulationsaktivität, verfahren zur isolierung und verwendungen Download PDF

Info

Publication number
EP1926813B2
EP1926813B2 EP06777197.2A EP06777197A EP1926813B2 EP 1926813 B2 EP1926813 B2 EP 1926813B2 EP 06777197 A EP06777197 A EP 06777197A EP 1926813 B2 EP1926813 B2 EP 1926813B2
Authority
EP
European Patent Office
Prior art keywords
cells
document
cell population
ifn
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP06777197.2A
Other languages
English (en)
French (fr)
Other versions
EP1926813B1 (de
EP1926813A2 (de
Inventor
Dirk Büscher
Manuel Ángel González de La Peña
Mario Delgado Mora
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Consejo Superior de Investigaciones Cientificas CSIC
Tigenix SA
Original Assignee
Consejo Superior de Investigaciones Cientificas CSIC
Tigenix SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=37708240&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP1926813(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to EP17154939.7A priority Critical patent/EP3184630B1/de
Priority to SI200632094T priority patent/SI1926813T2/sl
Priority to EP10195268.7A priority patent/EP2340847A3/de
Priority to EP17154940.5A priority patent/EP3184631B1/de
Priority to PL06777197T priority patent/PL1926813T5/pl
Application filed by Consejo Superior de Investigaciones Cientificas CSIC, Tigenix SA filed Critical Consejo Superior de Investigaciones Cientificas CSIC
Publication of EP1926813A2 publication Critical patent/EP1926813A2/de
Publication of EP1926813B1 publication Critical patent/EP1926813B1/de
Publication of EP1926813B2 publication Critical patent/EP1926813B2/de
Application granted granted Critical
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/35Fat tissue; Adipocytes; Stromal cells; Connective tissues
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/001Preparations to induce tolerance to non-self, e.g. prior to transplantation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46433Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0637Immunosuppressive T lymphocytes, e.g. regulatory T cells or Treg
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/122Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/24Interferons [IFN]

Definitions

  • the present invention relates to the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial utilizing cell populations derived from adult tissues.
  • the present invention provides a population of connective tissue derived cells that respond to interferon-gamma (IFN- ⁇ ) by expressing indolamine-2,3-dioxygenase (IDO) for use in preventing, treating or ameliorating one or more symptoms associated with Inflammatory Bowel Disease (IBD).
  • IFN- ⁇ interferon-gamma
  • IDO indolamine-2,3-dioxygenase
  • the immune system in higher vertebrates represents the first line of defence against various antigens that can enter the vertebrate body, including micro-organisms such as bacteria, fungi and viruses that are the causative agents of a variety of diseases.
  • the immune system is also involved in a variety of other diseases or disorders, including autoimmune or immunopathologic diseases, immunodeficiency syndromes, atherosclerosis and various neoplastic diseases.
  • Autoimmune diseases are caused when the body's immune system, which is meant to defend the body against bacteria, viruses, and any other foreign product, malfunctions and produces a pathological response against healthy tissue, cells and organs.
  • Antibodies, T cells and macrophages provide beneficial protection, but can also produce harmful or deadly immunological responses.
  • Autoimmune diseases can be organ specific or systemic and are provoked by different pathogenic mechanisms. Organ specific autoimmunization is characterized by aberrant expression of major-histocompatibility complex (MHC) antigens, antigenic mimicry and allelic variations in MHC genes.
  • Systemic autoimmune diseases involve polyclonal B cell activation and abnormalities of immunoregulatory T cells, T cell receptors and MHC genes. Examples of organ specific autoimmune diseases are diabetes, hyperthyroidism, autoimmune adrenal insufficiency, pure red cell anemia, multiple sclerosis and rheumatic carditis.
  • Representative systemic autoimmune diseases are systemic lupus erythematosus, chronic inflammation, Sjogren's syndrome, polymyositis, dermatomyositis and scleroderma.
  • Inflammation is a process by which the body's white blood cells and secreted factors protect our bodies from infection by foreign substances, such as bacteria and viruses.
  • Secreted factors known as cytokines and prostaglandins control this process, and are released in an ordered and self-limiting cascade into the blood or affected tissues.
  • IBD Inflammatory Bowel disease
  • IBD ulcerative colitis
  • CD is chronic and typically has an onset before 30 years of age, patients generally require lifelong treatment. Although its etiology remains unknown, there is circumstantial evidence to link CD to a failure of the mucosal immune system to attenuate the immune response to endogenous antigens.
  • Therapeutic agents currently used for CD including aminosalicylates, corticosteroids, azathioprine, 6-mercaptopurine, antibiotics, and methotrexate, are not entirely effective, nonspecific, and with multiple adverse side effects. In most cases, surgical resection is the ultimate alternative. Therefore, the present therapeutic strategy is to find drugs or agents that specifically modulate both components of the disease, i.e., the inflammatory and T-cells driven responses.
  • TNF anti-tumour necrosis factor
  • Infliximab the first treatment approved specifically for Crohn's disease, is an anti-tumour necrosis factor (TNF) antibody.
  • TNF is a protein produced by the immune system that may cause the inflammation associated with Crohn's disease.
  • Anti-TNF removes TNF from the bloodstream before it reaches the intestines, thereby preventing inflammation.
  • TNF is a very pleiotropic factor
  • severe side effects are relatively common, and its long-term safety is still to be determined.
  • the efficacy is also limited because many of the inflammatory processes that occur in the patients are not dependant on TNF signalling.
  • RA Rheumatoid arthritis
  • Rheumatoid arthritis and juvenile rheumatoid arthritis are types of inflammatory arthritis.
  • Arthritis is a general term that describes inflammation in joints. Some, but not all, types of arthritis are the result of misdirected inflammation.
  • Rheumatoid arthritis affects about 1% of the world's population and is essentially disabling.
  • Rheumatoid arthritis is an autoimmune disorder where the body's immune system improperly identifies the synovial membranes that secrete the lubricating fluid in the joints as foreign. Inflammation results, and the cartilage and tissues in and around the joints are damaged or destroyed. The body replaces damaged tissue with scar tissue, causing the normal spaces within the joints to become narrow and the bones to fuse together.
  • rheumatoid arthritis In rheumatoid arthritis, there is an autoimmune cycle of persistent antigen presentation, T-cell stimulation, cytokine secretion, synovial cell activation, and joint destruction.
  • the first line of treatment of any arthritis is usually anti-inflammatories, such as aspirin, ibuprofen and Cox-2 inhibitors such as celecoxib and rofecoxib.
  • Anti-TNF humanized monoclonal antibodies, such as Infliximab are also used; however, it has many secondary effects or side effects and its efficacy is quite low.
  • "Second line drugs” include gold, methotrexate and steroids.
  • T cells All immune responses are controlled by T cells.
  • Self-reactive cells with the potential to elicit autoimmune responses comprise a part of the normal T cell repertoire, but in the healthy state, their activation is prevented by suppressor cells.
  • suppressor cells were originally described in the 1970s, significant progress in characterizing T-cell subsets has been made only recently, when they have been renamed as regulatory T cells.
  • T-reg cells There are different CD4 + , CD8 + , natural killer cell, and ⁇ T cell subsets with regulatory (suppressor) activity.
  • Two major types of T-reg cells have been characterized in the CD4 + population, i.e., the naturally-occurring, thymus-generated T-reg cells, and the peripherally-induced, IL-10 or TGF- ⁇ secreting T-reg cells (Trl cells).
  • the CD4 + CD25 + , Foxp3-expressing, naturally-occurring T-reg cells generated in thymus migrate and are maintained in the periphery.
  • the signals for their thymic generation and maintenance in the periphery are not entirely defined, although both CD28 stimulation and IL-2 appear to be required.
  • CD4 + CD25 + T-reg cells in the periphery does not decrease with age, although these cells are anergic and prone to apoptosis, and their site of origin, the thymus, undergoes age-related involution. This suggests that the pool of CD4 + CD25 + T-reg cells is maintained peripherally.
  • Several experimental models support the idea of peripheral generation of CD4 + CD25 + T-reg cells from CD4 + CD25 - T cells.
  • the endogenous factors and mechanisms controlling the peripheral expansion of CD4 + CD25 + T-reg cells are mostly unknown.
  • TGF- ⁇ cytokine transforming growth factor-beta
  • IDO indoleamine 2,3-dioxygenase
  • IDO is expressed in some subsets of dendritic cells (DCs), which are key regulators of immune response (tolerogenic DCs). These DCs are able of suppressing in vivo T-cell responses by locally depleting tryptophan ( US Patent No. 2002/0155104 ).
  • DCs dendritic cells
  • tumour lines intestinal cells
  • trophoblasts express IDO.
  • the expression of IDO in trophoblasts appears to be constitutive and has been strongly correlated to tolerance of allogeneic tissue from the foetus. IDO is believed to induce apoptosis in T cells, and cause spontaneous tolerance to liver allografts.
  • IDO immunosuppressive activity
  • MSCs which are disclosed as being negative for CD14 and CD45 expression and positive for CD44 and CD105 expression, increase the regulatory T cell population when they are co-cultured with peripheral blood mononuclear cells (PBMCs) ( Aggarwal and Pittenger, Blood 2005; 105(4):1815-1822 ).
  • PBMCs peripheral blood mononuclear cells
  • the invention is based on the discovery that certain cell populations with multilineage potential which are present in different connective tissues are capable of acting as immunoregulatory agents in vivo and in vitro.
  • Inventors have isolated a population of connective tissue derived cells that respond to interferon-gamma (IFN- ⁇ ) by expressing indolamine-2,3-dioxygenase (IDO).
  • IFN- ⁇ interferon-gamma
  • IDO indolamine-2,3-dioxygenase
  • the immunoregulatory effects of said cells can be used for preventing, treating or ameliorating one or more symptoms associated with Inflammatory Bowel Disease (IBD).
  • IBD Inflammatory Bowel Disease
  • this document describes an isolated cell population from adipose tissue wherein the cells of said cell population: (i) do not express markers specific from antigen-presenting cells (APC) wherein the markers specific for APC are CD11b, CD11c, CD14, CD45 and HLAII; (ii) do not express indolamine 2,3-dioxygenase (IDO) constitutively; (iii) express IDO upon stimulation with interferon-gamma (IFN- ⁇ ); and (iv) present capacity to be differentiated into at least two cell lineages.
  • APC antigen-presenting cells
  • IDO indolamine 2,3-dioxygenase
  • IFN- ⁇ interferon-gamma
  • the document describes a method for the isolation of said cell population.
  • the cell population obtainable according to said method constitutes an additional disclosure of this document.
  • this invention relates to said cell population for use in the prevention, treatment or amelioration of one or more symptoms associated with Inflammatory Bowel Disease (IBD) in a subject suffering from IBD.
  • IBD Inflammatory Bowel Disease
  • a said inflammatory disease is a chronic inflammatory disease, such as, for example, Inflammatory Bowel Disease (IBD) or Rheumatoid Arthritis (RA).
  • IBD Inflammatory Bowel Disease
  • RA Rheumatoid Arthritis
  • the invention relates to the use of said cell population in the preparation of a medicament for preventing, treating or ameliorating one or more symptoms associated with Inflammatory Bowel Disease (IBD) in a subject suffering from IBD.
  • IBD Inflammatory Bowel Disease
  • the document relates to the use of said cell population in the in vitro preparation or generation of regulatory T-cells (T-reg).
  • T-reg regulatory T-cells
  • Said T-reg cell population as well as a method for the isolation thereof constitute further disclosures of this document.
  • the document also discloses said T-reg cell population for use as medicament, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating an inflammatory disease.
  • the document also discloses the use of said T-reg cell population in the preparation of a medicament, such as a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial, e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease, or a medicament for treating allergies, for example, but not limited to, hypersensitivity Type IV reactions.
  • a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease, or a medicament for treating allergies, for example, but not limited to, hypersensitivity Type IV reactions.
  • the document also discloses a method for the isolation of an irradiated cell population which comprises irradiating said cell population with a controlled source of ionizing radiation under appropriate conditions. Said irradiated cell population constitutes a further disclosure of this document.
  • the document also discloses said irradiated cell population for use as medicament, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating an inflammatory disease.
  • the document also discloses the use of said irradiated cell population in the preparation of a medicament, such as a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial, e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease.
  • a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease.
  • the document also discloses a method which comprises subjecting said cell population to treatment with interferon- ⁇ (IFN- ⁇ ). Said IFN- ⁇ -treated cell population constitutes a further disclosure of the document.
  • IFN- ⁇ interferon- ⁇
  • the document also discloses said IFN- ⁇ -treated cell population for use as medicament, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating an inflammatory disease.
  • the document also discloses the use of said IFN- ⁇ -treated cell population in the preparation of a medicament, such as a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial, e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease.
  • a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease.
  • the document also discloses a method which comprises subjecting said cell population to (i) irradiation, and (ii) stimulation with IFN- ⁇ , wherein treatments (i) and (ii) are carried out in any order.
  • Said irradiated IFN- ⁇ -pre-stimulated cell population or IFN- ⁇ -pre-stimulated irradiated cell population constitute a further disclosure of the document.
  • the document also discloses said irradiated IFN- ⁇ -pre-stimulated cell population or IFN- ⁇ -pre-stimulated irradiated cell population for use as medicament, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating an inflammatory disease.
  • the document also discloses the use of said irradiated IFN- ⁇ -pre-stimulated cell population or IFN- ⁇ -pre-stimulated irradiated cell population in the preparation of a medicament, such as a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial, e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease.
  • a medicament for the prevention, treatment or amelioration of one or more symptoms of disorders in which modulation of a subject's immune system is beneficial e.g., a medicament for inducing transplantation tolerance, or a medicament for treating autoimmune diseases, or a medicament for treating an inflammatory disease.
  • the document relates to the use of said cell population, or this document also discloses the use of said T-reg cell population, or said irradiated cell population, or said IFN- ⁇ -treated cell population, or said irradiated IFN- ⁇ -pre-stimulated cell population, or said IFN- ⁇ -pre-stimulated irradiated cell population for preventing, treating, or ameliorating one or more symptoms associated with autoimmune diseases, inflammatory disorders, or immunologically mediated diseases including rejection of transplanted organs and tissues.
  • the document discloses a method of preventing, treating, or ameliorating one or more symptoms associated with autoimmune diseases, inflammatory disorders, or immunologically mediated diseases, in a subject suffering from any of said disorders or diseases, which comprises administering to said subject in need of such treatment of a prophylactically or therapeutically effective amount of said cell population, or said T-reg cell population, or said irradiated cell population, or said IFN- ⁇ -treated cell population, or said irradiated IFN- ⁇ -pre-stimulated cell population, or said IFN- ⁇ -pre-stimulated irradiated cell population.
  • This document also discloses the use of such methods in combination therapy, in other words, a cell population disclosed by this document is coadministered with one or more agents, either simultaneously with the second or further agent, or separately, e.g., sequentially.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an isolated cell population from adipose tissue characterized in that the cells of said cell population: (i) do not express markers specific from antigen-presenting cells (APC) wherein the markers specific for APC are CD11b, CD11c, CD14, CD45 and HLAII; (ii) do not express indolamine 2,3-dioxygenase (IDO) constitutively; (iii) express IDO upon stimulation with interferon-gamma (IFN- ⁇ ); and (iv) present capacity to be differentiated into at least two cell lineages, and an acceptable pharmaceutically carrier, for use in preventing, treating, or ameliorating one or more symptoms associated with Inflammatory Bowel Disease (IBD) in a subject suffering from IBD.
  • APC antigen-presenting cells
  • IDO indolamine 2,3-dioxygenase
  • IFN- ⁇ interferon-gamma
  • the document discloses a method for distinguishing adult multipotent cells from differentiated cells comprising the step of verifying whether the cell expresses IDO upon stimulation with IFN- ⁇ .
  • the invention relates to a kit comprising an isolated cell population from adipose tissue characterized in that the cells of said cell population: (i) do not express markers specific from antigen-presenting cells (APC) wherein the markers specific for APC are CD11b, CD11c, CD14, CD45 and HLAII; (ii) do not express indolamine 2,3-dioxygenase (IDO) constitutively; (iii) express IDO upon stimulation with interferon-gamma (IFN- ⁇ ); and (iv) present capacity to be differentiated into at least two cell lineages, for use in preventing, treating, or ameliorating one or more symptoms associated with Inflammatory Bowel Disease (IBD) in a subject suffering from IBD.
  • APC antigen-presenting cells
  • IDO indolamine 2,3-dioxygenase
  • IFN- ⁇ interferon-gamma
  • immunosuppressant immunoregulatory effects of said cells can be used for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues.
  • APC antigen presenting cells
  • MHC major histocompatibility complex
  • autoimmune disease refers to a condition in a subject characterized by cellular, tissue and/or organ injury caused by an immunologic reaction of the subject to its own cells, tissues and/or organs.
  • autoimmune diseases which can be treated with the cell population disclosed by this document include alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, autoimmune Addison's disease, autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune oophoritis and orchitis, autoimmune thrombocytopenia, Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac sprue-dermatitis, chronic fatigue immune dysfunction syndrome (CFIDS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, cicatrical pemphigoid, CREST syndrome, cold agglutinin disease, discoid lupus, essential mixed
  • immunoregulatory agent refers to an agent that inhibits or reduces one or more biological activities of the immune system.
  • An immunoregulatory agent is an agent that inhibits or reduces one or more biological activities (e.g., the proliferation, differentiation, priming, effector function, production of cytokines or expression of antigens) of one or more immune cells (e.g., T cells).
  • inflammatory disease refers to a condition in a subject characterized by inflammation, e.g., chronic inflammation.
  • inflammatory disorders include, but are not limited to, rheumatoid arthritis (RA), Inflammatory Bowel Disease (IBD), asthma, encephalitis, chronic obstructive pulmonary disease (COPD), inflammatory osteolysis, allergic disorders, septic shock, pulmonary fibrosis (e.g., idiopathic pulmonary fibrosis), inflammatory vaculitides (e.g., polyarteritis nodosa, Wegner's granulomatosis, Takayasu's arteritis, temporal arteritis, and lymphomatoid granulomatosus), post-traumatic vascular angioplasty (e.g., restenosis after angioplasty), undifferentiated spondyloarthropathy, undifferentiated arthropathy, arthritis, inflammatory osteolysis, chronic hepatitis
  • RA rheumato
  • isolated applied to a cell population refers to a cell population, isolated from the human or animal body, which is substantially free of one or more cell populations that are associated with said cell population in vivo or in vitro.
  • MHC major histocompatibility complex
  • HLA human leukocyte antigen
  • subject refers to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, or mouse) and a primate (e.g., a monkey, or a human). In a preferred embodiment, the subject is a human.
  • a non-primate e.g., a cow, pig, horse, cat, dog, rat, or mouse
  • a primate e.g., a monkey, or a human.
  • the subject is a human.
  • T-cell refers to cells of the immune system which are a subset of lymphocytes that express the T cell receptor (TCR).
  • T-reg cells refers to T cell subsets that actively suppress activation of the immune system and prevent pathological self-reactivity, i.e. an autoimmune disease.
  • the terms “treat”, “treatment” and “treating” refer to the amelioration of one or more symptoms associated with a disorder including, but not limited to, an inflammatory disorder, an autoimmune disease or an immunologically mediated disease including rejection of transplanted organs and tissues, that results from the administration of the cell population disclosed by the document, the T-reg cell population disclosed by the document, or the IFN- ⁇ -pre-stimulated cell population disclosed by the document, or a pharmaceutical composition comprising same, to a subject in need of said treatment.
  • a disorder including, but not limited to, an inflammatory disorder, an autoimmune disease or an immunologically mediated disease including rejection of transplanted organs and tissues, that results from the administration of the cell population disclosed by the document, the T-reg cell population disclosed by the document, or the IFN- ⁇ -pre-stimulated cell population disclosed by the document, or a pharmaceutical composition comprising same, to a subject in need of said treatment.
  • combination therapy refers to the use of the cell populations disclosed by the present document with other active agents or treatment modalities, in the manner of the present document for the amelioration of one or more symptoms associated with a disorder including, but not limited to, an inflammatory disorder, an autoimmune disease or an immunologically mediated disease including rejection of transplanted organs and tissues.
  • agents or treatments may include known drugs and therapies for the treatment of such disorders.
  • the cell populations disclosed by this document may also be combined with corticosteroids, non-steroidal anti-inflammatory compounds, or other agents useful in treating inflammation.
  • the combined use of the agents of the present document with these other therapies or treatment modalities may be concurrent, or given sequentially, that is, the two treatments may be divided up such that a cell population or a pharmaceutical composition comprising same of the present document may be given prior to or after the other therapy or treatment modality.
  • the attending physician may decide on the appropriate sequence of administering the cell population, or a pharmaceutical composition comprising same, in combination with other agents, therapy or treatment modality.
  • the present document discloses an isolated cell population from connective tissue, hereinafter referred to as "cell population disclosed by the document”, characterised in that the cells of said cell population:
  • connective tissue refers to tissue derived from mesenchyme and includes several tissues which are characterized in that their cells are included within the extracellular matrix. Among the different types of connective tissues, adipose and cartilaginous tissues are included.
  • the cells disclosed by this document are from the stromal fraction of the adipose tissue.
  • the cells disclosed by this document are obtained from chondrocytes, the only cells found in the hyaline cartilage.
  • the cells disclosed by this document are obtained from skin. Also, in another particular disclosure, the cells disclosed by this document are obtained from bone marrow.
  • the cells disclosed by this document can be obtained from any suitable source of connective tissue from any suitable animal, including humans. In general, said cells are obtained from non-pathological post-natal mammalian connective tissues. In a preferred disclosure, the cells disclosed by this document are obtained from a source of connective tissue, such as the stromal fraction of adipose tissue, hyaline cartilage, bone marrow, skin etc. Also, in a particular disclosure, the cells of the cell population disclosed by this document are from a mammal, e.g., a rodent, primate, etc., preferably, from a human.
  • a mammal e.g., a rodent, primate, etc.
  • the cells disclosed by this document are characterized in that (i) they do not express markers specific from APCs; (ii) they do not express IDO constitutively; (iii) they express IDO upon stimulation with IFN- ⁇ ; and (iv) they present capacity to be differentiated into at least two cell lineages.
  • the cells disclosed by this document are negative for at least one, two, three, four or preferably all of the following markers CD11b, CD11c, CD14, CD45, and HLAII, which are specific markers for APCs lineages. Thus, the cells disclosed by this document do not constitute a previously described subpopulation of specialized APCs.
  • the cells disclosed by this document are negative for at least one, two of, or preferably all of the following cell surface markers: CD31, CD34 and CD133.
  • negative with respect to cell surface markers means that, in a cell population comprising the cells disclosed by this document , less than 10%, preferably 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1% or none of the cells show a signal for a specific cell surface marker in flow cytometry above the background signal, using conventional methods and apparatus (for example .a Beckman Coulter Epics XL FACS system used with commercially available antibodies and standard protocols known in the art).
  • the cells disclosed by this document are characterised in that they express at least one, two, three, four, of or preferably all of the following cell surface markers: CD9, CD44, CD54, CD90 and CD105; i.e., the cells disclosed by this document are positive for at least one, two, three, four of and preferably all said cell surface markers (CD9, CD44, CD54, CD90 and CD105).
  • the cells disclosed by this document are characterised in that they have significant expression levels of at least one, two, three, four, of and preferably all of said cell surface markers (CD9, CD44, CD54, CD90 and CD105).
  • the expression "significant expression” means that, in a cell population comprising the cells disclosed by this document , more than 10%, preferably 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or all of the cells show a signal for a specific cell surface marker in flow cytometry above the background signal using conventional methods and apparatus (for example .a Beckman Coulter Epics XL FACS system used with commercially available antibodies and standard protocols known in the art).
  • the background signal is defined as the signal intensity given by a nonspecific antibody of the same isotype as the specific antibody used to detect each surface marker in conventional FACS analysis.
  • the specific signal observed is stronger than 10%, preferably 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 500%, 1000%, 5000%, 10000% or above, than the background signal intensity using conventional methods and apparatus (for example .a Beckman Coulter Epics XL FACS system used with commercially available antibodies and standard protocols known in the art).
  • the cells disclosed by this document are also negative for the cell surface marker CD106(VCAM-1),.
  • examples of such cells are certain populations of adipose tissue-derived stromal stem cells as described herein.
  • the cells disclosed by this document do not express IDO constitutively, but they express IDO upon stimulation with IFN- ⁇ .
  • IFN- ⁇ IL-1
  • IL-1 interleukin-1
  • TNF- ⁇ tumour necrosis factor-alpha
  • endotoxin LPS used at a concentration of 100ng/ml
  • Stimulation with IFN- ⁇ for example at 3ng/ml or higher can also induce expression of HLAII in the cells disclosed by this document to give a positive signal as defined herein for a cell surface marker.
  • Said expression can be detected by those skilled in the art using any known technique that allows the detection of the expression of specific proteins.
  • said techniques are cell cytometry techniques.
  • the cells disclosed by this document present the capacity to proliferate and be differentiated into at least two, more preferably three, four, five, six, seven or more cell lineages.
  • Illustrative, non-limiting examples of cell lineages in which the cells disclosed by this document can be differentiated include osteocytes, adipocytes, chondrocytes, tenocytes, myocytes, cardiomyocytes, hematopoietic-supporting stromal cells, endothelial cells, neurons, astrocytes, and hepatocytes.
  • Cells disclosed by this document can proliferate and differentiate into cells of other lineages by conventional methods. Methods of identifying and subsequently isolating differentiated cells from their undifferentiated counterparts can be also carried out by methods well known in the art.
  • the cells disclosed by this document are also capable of being expanded ex vivo. That is, after isolation, the cells disclosed by this document can be maintained and allowed to proliferate ex vivo in culture medium.
  • culture medium is composed of, for example, Dulbecco's Modified Eagle's Medium (DMEM), with antibiotics (for example, 100units/ml Penicillin and 100 ⁇ g/ml Streptomycin) or without antibiotics, and 2 mM glutamine, and supplemented with 2-20% fetal bovine serum (FBS). It is within the skill of one in the art to modify or modulate concentrations of media and/or media supplements as necessary for the cells used. Sera often contain cellular and non-cellular factors and components that are necessary for viability and expansion.
  • DMEM Dulbecco's Modified Eagle's Medium
  • FBS fetal bovine serum
  • sera examples include FBS, bovine serum (BS), calf serum (CS), fetal calf serum (FCS), newborn calf serum (NCS), goat serum (GS), horse serum (HS), porcine serum, sheep serum, rabbit serum, rat serum (RS), etc.
  • BS bovine serum
  • CS calf serum
  • FCS fetal calf serum
  • NCS newborn calf serum
  • GS goat serum
  • HS horse serum
  • porcine serum sheep serum
  • rabbit serum rat serum
  • cells disclosed by this document will benefit from FBS concentrations of about 2% to about 25%.
  • the cells disclosed by this document can be expanded in a culture medium of definite composition, in which the serum is replaced by a combination of serum albumin, serum transferrin, selenium, and recombinant proteins including but not limited to: insulin, platelet-derived growth factor (PDGF), and basic fibroblast growth factor (bFGF) as known in the art.
  • PDGF platelet-derived growth factor
  • bFGF basic fibroblast growth factor
  • amino acids include, but are not limited to, L-alanine, L- arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine, and the like.
  • Antimicrobial agents are also typically used in cell culture to mitigate bacterial, mycoplasmal, and fungal contamination.
  • antibiotics or anti-mycotic compounds used are mixtures of penicillin/streptomycin, but can also include, but are not limited to amphotericin (Fungizone®), ampicillin, gentamicin, bleomycin, hygromacin, kanamycin, mitomycin, etc.
  • Hormones can also be advantageously used in cell culture and include, but are not limited to, D-aldosterone, diethylstilbestrol (DES), dexamethasone, b-estradiol, hydrocortisone, insulin, prolactin, progesterone, somatostatin/human growth hormone (HGH), etc.
  • DES diethylstilbestrol
  • HGH somatostatin/human growth hormone
  • the maintenance conditions of the cells disclosed by this document can also contain cellular factors that allow cells to remain in an undifferentiated form. It is apparent to those skilled in the art that prior to differentiation, supplements that inhibit cell differentiation must be removed from the culture medium. It is also apparent that not all cells will require these factors. In fact, these factors may elicit unwanted effects, depending on the cell type.
  • the cells disclosed by this document lack in vivo tumorigenic activity.
  • said cells are characterized in that they do not present tumorigenic activity, i.e., they do not present an altered behaviour or proliferative phenotype which gives rise to a tumour cell
  • the cells disclosed by this document can be administered to a subject suffering from autoimmune diseases, inflammatory diseases or immunologically mediated diseases, such as rejection of transplanted organs and tissues, for suppressing the immune response. Thus, it is necessary that the cells disclosed by this document do not present tumorigenic activity.
  • the tumorigenic activity of the cells disclosed by this document can be tested by performing animal studies using immunodeficient mice strains. In these experiments, several million cells are implanted subcutaneously in the recipient animals, which are maintained for several weeks and analyzed for tumour formation. A particular assay is disclosed in Example 3.
  • the cells disclosed by this document can be transfected or genetically engineered to express, at least, one antigenic polypeptide.
  • the antigen comprises a purified or a synthetic or recombinant polypeptide representing a specific antigen to which it is desired that tolerance is to be induced, or a short synthetic polypeptide fragment derived from the amino acid sequence of such an antigen.
  • the source of antigen comprises antigens expressed by a donor tissue graft.
  • the source of antigen comprises a protein to which a patient has an autoimmune disorder.
  • the present document discloses a method for isolating a cell population from connective tissue, wherein the cells of said cell population present a phenotype characterized in that (i) they do not express markers specific from APCs; (ii) they do not express IDO constitutively; (iii) they express IDO upon stimulation with IFN- ⁇ ; and (iv) they present capacity to be differentiated into at least two cell lineages, said method comprising the steps of:
  • solid surface refers to any material that allows the cells disclosed by this document to adhere.
  • said material is a plastic material treated to promote the adhesion of mammalian cells to its surface, for example commercially available polystyrene plates optionally coated with poly-D-Lysine or other reagents.
  • Steps (i)-(vi) can be carried out by conventional techniques known by those skilled in the art.
  • the cells disclosed by this document can be obtained by conventional means from any suitable source of connective tissue from any suitable animal, including humans, e.g., from human adipose tissue or cartilaginous tissue.
  • the animal can be alive or dead, so long as connective tissue cells within the animal are viable.
  • human adipose cells are obtained from living donors, using well-recognized protocols such as surgical or suction lipectomy. Indeed, as liposuction procedures are so common, liposuction effluent is a particularly preferred source from which the cells disclosed by this document can be derived.
  • the cells disclosed by this document are from the stromal fraction of human adipose tissue obtained by liposuction.
  • the cells disclosed by this document are from human hyaline articular cartilage obtained by arthroscopic techniques.
  • the cells disclosed by this document are from human skin obtained by biopsy techniques.
  • the cells disclosed by this document are from human bone marrow obtained by aspiration.
  • the sample of connective tissue is, preferably, washed before being processed to separate the cells disclosed by this document from the remainder of the material.
  • the sample of connective tissue is washed with physiologically-compatible saline solution (e.g., phosphate buffered saline (PBS)) and then vigorously agitated and left to settle, a step that removes loose matter (e.g., damaged tissue, blood, erythrocytes, etc) from the tissue.
  • physiologically-compatible saline solution e.g., phosphate buffered saline (PBS)
  • PBS phosphate buffered saline
  • the washing and settling steps generally are repeated until the supernatant is relatively clear of debris.
  • the remaining cells generally will be present in clumps of various sizes, and the protocol proceeds using steps gauged to degrade the gross structure while minimizing damage to the cells themselves.
  • One method of achieving this end is to treat the washed lumps of cells with an enzyme that weakens or destroys bonds between cells (e.g., collagenase, dispase, trypsin, etc.).
  • an enzyme that weakens or destroys bonds between cells e.g., collagenase, dispase, trypsin, etc.
  • the amount and duration of such enzymatic treatment will vary, depending on the conditions employed, but the use of such enzymes is generally known in the art.
  • the lumps of cells can be degraded using other treatments, such as mechanical agitation, sonic energy, thermal energy, etc. If degradation is accomplished by enzymatic methods, it is desirable to neutralize the enzyme following a suitable period, to minimize deleterious effects on the cells.
  • the degradation step typically produces a slurry or suspension of aggregated cells and a fluid fraction containing generally free stromal cells (e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells).
  • stromal cells e.g., red blood cells, smooth muscle cells, endothelial cells, fibroblast cells, and stem cells.
  • the next stage in the separation process is to separate the aggregated cells from the cells disclosed by this document. This can be accomplished by centrifugation, which forces the cells into a pellet covered by a supernatant. The supernatant then can be discarded and the pellet suspended in a physiologically-compatible fluid.
  • the suspended cells typically include erythrocytes, and in most protocols it is desirable to lyse them.
  • erythrocytes Methods for selectively lysing erythrocytes are known in the art, and any suitable protocol can be employed (e.g., incubation in a hyper -or hypotonic medium, by lysis using ammonium chloride, etc.). Of course, if the erythrocytes are lysed, the remaining cells should then be separated from the lysate, for example by filtration, sedimentation, or density fractionation.
  • the suspended cells can be washed, re-centrifuged, and resuspended one or more successive times to achieve greater purity.
  • the cells can be separated on the basis of cell surface marker profile or on the basis of cell size and granularity.
  • the cells can be cultured and, if desired, assayed for number and viability to assess the yield.
  • the cells will be cultured without differentiation, on a solid surface, using a suitable cell culture media, at the appropriate cell densities and culture conditions.
  • a suitable cell culture medium e.g., DMEM, typically supplemented with 5-15% (e.g., 10%) of a suitable serum, such as fetal bovine serum or human serum
  • cells are washed in order to remove non-adhered cells and cell fragments.
  • the cells are maintained in culture in the same medium and under the same conditions until they reach the adequate confluence, typically, about 80% cell confluence, with replacement of the cell culture medium when necessary.
  • the cells can be expanded by means of consecutive passages using a detachment agent such as trypsin and seeding onto a bigger cell culture surface at the appropriate cell density (usually 2,000-10,000 cells/cm 2 ).
  • cells are then passaged at least two times in such medium without differentiating, while still retaining their developmental phenotype, and more preferably, the cells can be passaged at least 10 times (e.g., at least 15 times or even at least 20 times) without losing developmental phenotype.
  • the cells are plated at a desired density such as between about 100 cells/cm 2 to about 100,000 cells/cm 2 (such as about 500 cells/cm 2 to about 50,000 cells/cm 2 , or, more particularly, between about 1,000 cells/cm 2 to about 20,000 cells/cm 2 ). If plated at lower densities (e.g., about 300 cells/cm 2 ), the cells can be more easily clonally isolated.
  • the cell density is between 2,000-10,000 cells/cm 2 .
  • Cells which remain adhered to the solid surface after such treatment comprising at least two passages are selected and the phenotype of interest is analyzed by conventional methods in order to confirm the identity of the cells disclosed by this document as will be mentioned below.
  • Cells which remain adhered to the solid surface after the first passage are from heterogeneous origin; therefore, said cells must be subjected to at least another passage.
  • Example 1 describes in a detailed manner the isolation of the cells disclosed by this document from human adipose tissue and from human cartilaginous tissue.
  • Cell-surface markers can be identified by any suitable conventional technique, usually based on a positive/negative selection; for example, monoclonal antibodies against cell-surface markers, whose presence/absence in the cells has to be confirmed, can be used; although other techniques can also be used.
  • monoclonal antibodies against one, two, three, four, five, six, seven of or preferably all of CD11b, CD11c, CD14, CD45, HLAII, CD31, CD34 and CD133 are used in order to confirm the absence of said markers in the selected cells; and monoclonal antibodies against one, two, three, four, of or preferably all of CD9, CD44, CD54, CD90 and CD105 are used in order to confirm the presence thereof or detectable expression levels of, at least one of and preferably all of, said markers.
  • Said monoclonal antibodies are known, commercially available or can be obtained by a skilled person in the art by conventional methods.
  • IFN- ⁇ -inducible IDO activity in the selected cells can be determined by any suitable conventional assay.
  • the selected cells can be stimulated with IFN- ⁇ and assayed for IDO expression; then conventional Western-blot analysis for IDO protein expression can be performed and IDO enzyme activity following IFN- ⁇ stimulation of the selected cells can be measured by tryptophan-to-kynurenine conversion with for example via High Performance Liquid Chromatography (HPLC) analysis and photometric determination of kynurenine concentration in the supernatant as the readout.
  • HPLC High Performance Liquid Chromatography
  • Example 2 A suitable assay for determining IFN- ⁇ -inducible IDO activity in the selected cells is disclosed in Example 2.
  • the amount of IDO produced depends on the number of cells per square centimetre, which is preferably at a level of 5000cells/cm 2 or more, but not limited to this concentration and the concentration of IFN- ⁇ , which ideally is 3ng/ml or more, but not limited to this concentration.
  • the activity of IDO produced under the described conditions will result in a detectable production of kynurenine in the ⁇ M range after 24hours or more.
  • the capacity of the selected cells to differentiate into at least two cell lineages can be assayed by conventional methods as known in the art.
  • the cells and cell populations described by the instant document can be clonally expanded, if desired, using a suitable method for cloning cell populations.
  • a proliferated population of cells can be physically picked and seeded into a separate plate (or the well of a multi-well plate).
  • the cells can be subcloned onto a multi-well plate at a statistical ratio for facilitating placing a single cell into each well (e.g., from about 0.1 to about 1 cell/well or even about 0.25 to about 0.5 cells/well, such as 0.5 cells/well).
  • the cells can be cloned by plating them at low density (e.g., in a Petri dish or other suitable substrate) and isolating them from other cells using devices such as a cloning rings.
  • the production of a clonal population can be expanded in any suitable culture medium.
  • the isolated cells can be cultured to a suitable point when their developmental phenotype can be assessed.
  • ex vivo expansion of the cells disclosed by this document without inducing differentiation can be accomplished for extended time periods for example by using specially screened lots of suitable serum (such as fetal bovine serum or human serum). Methods for measuring viability and yield are known in the art (e. g., trypan blue exclusion).
  • any of the steps and procedures for isolating the cells of the cell population disclosed by this document can be performed manually, if desired.
  • the process of isolating such cells can be facilitated and/or automated through one or more suitable devices, examples of which are known in the art.
  • the cells disclosed by this document can be used for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and rejection of transplanted organs and tissues.
  • the document relates to an isolated cell population from adipose tissue characterised in that the cells of said cell population:
  • the terms “disorder” and “disease” are used interchangeably to refer to a condition in a subject.
  • autoimmune disease is used interchangeably with the term “autoimmune disorder” to refer to a condition in a subject characterized by cellular, tissue and/or organ injury caused by an immunologic reaction of the subject to its own cells, tissues and/or organs.
  • inflammatory disease is used interchangeably with the term “inflammatory disorder” to refer to a condition in a subject characterized by inflammation, preferably chronic inflammation.
  • Autoimmune disorders may or may not be associated with inflammation.
  • inflammation may or may not be caused by an autoimmune disorder.
  • certain disorders may be characterized as both autoimmune and inflammatory disorders.
  • any autoimmune disease, inflammatory disorder or immunological mediated disease can be treated with the cells disclosed by this document.
  • Illustrative, non-limiting examples of said diseases and disorders which can be treated are those previously listed under heading "Definitions”.
  • said inflammatory disease is a chronic inflammatory disease, such as, e.g., IBD or RA.
  • the present invention relates to the use of an isolated cell population from adipose tissue characterised in that the cells of said cell population
  • the present document relates to the use of an isolated cell population from adipose tissue characterised in that the cells of said cell population
  • T-reg regulatory T-cells
  • cells including Foxp3+CD4+CD25+ T-reg and IL-10/TGFb-producing Tr1 cells
  • pathological self-reactivity i.e. an autoimmune disease, obtainable from the cells disclosed by the document, hereinafter referred to T-reg cells disclosed by the document.
  • the present document relates to a method for the isolation of a T-reg cell population disclosed by the document, which comprises:
  • the cells disclosed by this document can be used to produce a subset of T-cells, the T-reg cells disclosed by this document, which constitutes an additional disclosure of the present document.
  • the T-reg cells disclosed by this document can be isolated by conventional means known by a skilled person in the art.
  • T-reg cells disclosed by this document can be used for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues. Said use constitutes an additional disclosure of the present document.
  • the T-reg cells disclosed by this document are used as a medicament.
  • medicaments containing the T-reg cells disclosed by this document may be used for inducing transplantation tolerance, or for treating, and thereby alleviating, symptoms of autoimmune or inflammatory disorders, or immunologically mediated diseases including rejection of transplanted organs and tissues, in a subject suffering from any of said disorders or diseases.
  • the T-reg cells disclosed by this document can be used to therapeutically or prophylactically treat and thereby alleviating symptoms of autoimmune or inflammatory disorders in a subject suffering from any of said disorders or to alleviate symptoms of immunologically mediated diseases in a subject suffering from said diseases.
  • any autoimmune disease, inflammatory disorder or immunological mediated disease can be treated with the T-reg cells disclosed by this document.
  • Illustrative, non-limiting examples of said diseases and disorders which can be treated are those previously listed under heading "Definitions”.
  • said inflammatory disease is a chronic inflammatory disease, such as, e.g., IBD or RA.
  • the present document discloses the use of the T-reg cells disclosed by this document for the preparation of a medicament for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues.
  • this document further describes the use of the T-reg cells disclosed by this document for the preparation of a medicament for suppressing the immune response, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating inflammatory disorders. Examples of said autoimmune diseases and inflammatory diseases have been previously mentioned.
  • disease is an inflammatory disease, such as a chronic inflammatory disease, e.g., IBD or RA.
  • This document also discloses the use of cell populations disclosed by this document in the production of Treg cells specific for a chosen antigen or group of antigens and the use of these in the treatment of disease or disorders relating to that antigen or group of antigens.
  • antigens are those that play a role in autoimmune diseases, such as, for example, rheumatoid arthritis, Crohn's disease, hypersensitivity reaction Type IV, lupus, psoriasis and other autoimmune disorders known in the art and described elsewhere herein.
  • cell populations disclosed by this document are cultured in vitro in the presence of a chosen antigen, group of antigens or cell types expressing and/or presenting this antigen or antigens.
  • the cells disclosed by this document can optionally be prestimulated with IFN ⁇ , LPS or other activating agents known in the art. After a culture period of about 2, 4, 6, 12, 24, 48 or more hours, preferably between about 12 to about 24 hours, the cell population disclosed by this document is further co-cultured, optionally after the removal of the antigen, group of antigens or cells carrying said antigen, with peripheral blood leukocytes obtained from a subject. This co-culturing will result in the production of Treg cells specific for the chosen antigen, which can be used for treatment of the subject. Optionally these Treg cells can be expanded in number ex vivo using culture techniques known in the art before being administered to the patient.
  • the Inventors believe that the cell populations disclosed by this document are capable of presenting the chosen antigen via HLA Class II on the cell surface (seeming induced by IFN ⁇ ) to the peripheral blood leukocytes such that Treg cells are augmented and or activated within the population of peripheral blood leukocytes.
  • the Inventors have demonstrated that cell populations disclosed by this document are able to phagocytose small molecular weight molecules and thus are capable of presenting such molecules after IFN ⁇ stimulation via HLA Class II molecules.
  • the presentation of chosen antigen via this mechanism with the interaction with the peripheral blood leukocytes is believed to result in the above described Treg cell production.
  • a cell population disclosed by this document is administered directly in vivo without any co-culturing and can generate specific Treg cells, which in turn can treat a disorder.
  • Treg cells specific for a chosen antigen or group of antigens which comprises:
  • This document also describes the use of the specific Treg cells of step (c) in the treatment of diseases and disorders related to said chosen antigen or groups of antigens by administration of said Treg cells to the subject from which the peripheral blood leukocytes were obtained.
  • the cell population disclosed by this document as used in this method may be from the subject (autologous) or from a donor (allogeneic).
  • the cells disclosed by this document can be irradiated using a suitable controlled source of ionizing radiation, such a gamma irradiator device.
  • a suitable controlled source of ionizing radiation such as a gamma irradiator device.
  • the irradiation conditions must be experimentally adjusted by a person skilled in the art to determine the required exposure time to impart a radiation dose that cause the long term growth arrest of the cells disclosed by this document.
  • Said radiation dose can be for example 1-100, 5-85, 10-70, 12-60 Gy or more preferably15-45 Gy.
  • irradiation of the cells disclosed by this document before administration to the subject may result beneficial since said irradiation treatment makes cells incapable to proliferate or survive for long time periods in the subject.
  • Said irradiated cells constitute a further disclosure of the instant document.
  • the irradiated cells disclosed by this document can be used for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues. Said use constitutes an additional disclosure of the present document.
  • the irradiated cells disclosed by this document are used as a medicament.
  • medicaments containing the irradiated cells disclosed by this document may be used for inducing transplantation tolerance, or for treating, and thereby alleviating, symptoms of autoimmune or inflammatory disorders, or immunologically mediated diseases including rejection of transplanted organs and tissues, in a subject suffering from any of said disorders or diseases.
  • the irradiated cells disclosed by this document can be used to therapeutically or prophylactically treat and thereby alleviating symptoms of autoimmune or inflammatory disorders in a subject suffering from any of said disorders or to alleviate symptoms of immunologically mediated diseases in a subject suffering from said diseases.
  • any autoimmune disease, inflammatory disorder or immunological mediated disease can be treated with the irradiated cells disclosed by this document.
  • Illustrative, non-limiting examples of said diseases and disorders which can be treated are those previously listed under heading "Definitions”.
  • said inflammatory disease is a chronic inflammatory disease, such as, e.g., IBD or RA.
  • the present document relates to the use of the irradiated cells disclosed by this document for the preparation of a medicament for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues.
  • this document further describes the use of the irradiated cells disclosed by this document for the preparation of a medicament for suppressing the immune response, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating inflammatory disorders. Examples of said autoimmune diseases and inflammatory diseases have been previously mentioned.
  • disease is an inflammatory disease, such as a chronic inflammatory disease, e.g., IBD or RA.
  • the cells disclosed by this document can be pre-stimulated with IFN- ⁇ .
  • the methods for pre-stimulation with IFN- ⁇ are evident to those skilled in the art, and a procedure is given in Example 2.
  • the cells are pre-stimulated using a concentration of IFN- ⁇ between 0.1 and 100, 0.5 and 85, 1 and 70, 1.5 and 50, 2.5 and 40 ng/ml or more preferably 3 and 30 ng/ml, and a stimulation time preferably longer than 12 hours, for example, 13, 18, 24, 48, 72 hours or more.
  • pre-stimulation of the cells disclosed by this document with IFN- ⁇ before administration to the subject may result beneficial since the time period between IFN- ⁇ -pre-stimulated cell administration and IDO expression in the subject can be reduced.
  • the present document describes a method which comprises the treatment of the cells disclosed by this document with IFN- ⁇ in order to pre-stimulate said cells.
  • the cells obtainable according to said method hereinafter referred to "IFN- ⁇ -pre-stimulated cells disclosed by this document, constitutes an additional disclosure of the present document.
  • IFN- ⁇ -pre-stimulated cells disclosed by this document can be isolated by conventional means known by a skilled person in the art.
  • the IFN- ⁇ -pre-stimulated cells disclosed by this document can be used for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues. Said use constitutes an additional disclosure of the present document.
  • the IFN- ⁇ -pre-stimulated cells disclosed by this document are used as a medicament.
  • medicaments containing the IFN- ⁇ -pre-stimulated cells disclosed by this document may be used for inducing transplantation tolerance, or for treating, and thereby alleviating, symptoms of autoimmune or inflammatory disorders, or immunologically mediated diseases including rejection of transplanted organs and tissues, in a subject suffering from any of said disorders or diseases.
  • the IFN- ⁇ -pre-stimulated cells disclosed by this document can be used to therapeutically or prophylactically treat and thereby alleviating symptoms of autoimmune or inflammatory disorders in a subject suffering from any of said disorders or to alleviate symptoms of immunologically mediated diseases in a subject suffering from said diseases.
  • any autoimmune disease, inflammatory disorder or immunological mediated disease can be treated with the IFN- ⁇ -pre-stimulated cells disclosed by this document.
  • Illustrative, non-limiting examples of said diseases and disorders which can be treated are those previously listed under heading "Definitions”.
  • said inflammatory disease is a chronic inflammatory disease, such as, e.g., IBD or RA.
  • the present document describes the use of the IFN- ⁇ -pre-stimulated cells disclosed by this document for the preparation of a medicament for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues.
  • this document further describes the use of the IFN- ⁇ -pre-stimulated cells disclosed by this document for the preparation of a medicament for suppressing the immune response, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating inflammatory disorders. Examples of said autoimmune diseases and inflammatory diseases have been previously mentioned.
  • disease is an inflammatory disease, such as a chronic inflammatory disease, e.g., IBD or RA.
  • the cells disclosed by this document can be subjected to the treatments of irradiation and IFN- ⁇ -stimulation, in any order; i.e., cells disclosed by this document can be subjected firstly to irradiation and the resulting cells can be subsequently subjected to IFN- ⁇ -stimulation, or vice versa, cells disclosed by this document can be subjected firstly to IFN- ⁇ -stimulation and subsequently the resulting cells can be subjected to irradiation.
  • the cells disclosed by this document can be pre-stimulated with IFN- ⁇ and the resulting cells (IFN- ⁇ -pre-stimulated cells disclosed by the document) can be irradiated to render irradiated cells hereinafter referred to as "irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document”.
  • the cells disclosed by this document can be irradiated and the resulting cells (irradiated cells disclosed by this document) can be pre-stimulated with IFN- ⁇ to render IFN- ⁇ -prestimulated cells hereinafter referred to as "IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document”.
  • the present document refers to a method which comprises subjecting the cells disclosed by this document to (i) irradiation, and (ii) stimulation with IFN- ⁇ , wherein treatments (i) and (ii) can be carried out in any order, in order to irradiate IFN- ⁇ -pre-stimulated cells or to INF- ⁇ -pre-stimulate irradiated cells.
  • the cells obtainable according to said method herein referred to as "irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document or "IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document", respectively, constitutes additional disclosures of the present document.
  • Said irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document as well as said IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document can be isolated by conventional means known by a skilled person in the art.
  • administration to a subject of the cells disclosed by this document previously subjected to irradiation and IFN- ⁇ -stimulation may result beneficial for the reasons previously mentioned (e.g., subjecting cells to an irradiation treatment to make the cells incapable of proliferating or surviving for long time periods in the subject, whereas pre-stimulation of cells with IFN- ⁇ before administration to the subject may involve a reduction in the time period between IFN- ⁇ -pre-stimulated cell administration and IDO expression in the subject.
  • the irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document as well as the IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document can be used for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues. Said use constitutes an additional disclosure of the present document.
  • the irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document as well as the IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document are used as a medicament.
  • medicaments containing the irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document or the IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document may be used for inducing transplantation tolerance, or for treating, and thereby alleviating, symptoms of autoimmune or inflammatory disorders, or immunologically mediated diseases including rejection of transplanted organs and tissues, in a subject suffering from any of said disorders or diseases.
  • the irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document as well as the IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document can be used to therapeutically or prophylactically treat and thereby alleviating symptoms of autoimmune or inflammatory disorders in a subject suffering from any of said disorders or to alleviate symptoms of immunologically mediated diseases in a subject suffering from said diseases.
  • any autoimmune disease, inflammatory disorder or immunological mediated disease can be treated with the irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document or with the IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document.
  • Illustrative, non-limiting examples of said diseases and disorders which can be treated are those previously listed under heading "Definitions”.
  • said inflammatory disease is a chronic inflammatory disease, such as, e.g., IBD or RA.
  • the present document describes the use of the irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document or the IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document for the preparation of a medicament for preventing, treating or ameliorating one or more symptoms associated with disorders in which modulation of a subject's immune system is beneficial, including, but not limited to, autoimmune diseases, inflammatory disorders, and immunologically mediated diseases including rejection of transplanted organs and tissues.
  • this document further refers to the use of the irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document or the IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document for the preparation of a medicament for suppressing the immune response, or for inducing transplantation tolerance, or for treating autoimmune diseases, or for treating inflammatory disorders.
  • autoimmune diseases and inflammatory diseases have been previously mentioned.
  • disease is an inflammatory disease, such as a chronic inflammatory disease, e.g., IBD or RA.
  • the present invention provides pharmaceutical compositions for the treatment, prophylaxis, and amelioration of one or more symptoms associated with a disorder in which modulation of a subject's immune system is beneficial such as autoimmune diseases, inflammatory disorders, and rejection of transplanted organs and tissues.
  • the invention relates to a pharmaceutical composition, hereinafter referred to as the pharmaceutical composition of the invention, comprising an isolated cell population from adipose tissue characterised in that the cells of said cell population
  • the pharmaceutical composition disclosed herein comprises a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic agents (i.e., cell disclosed by the document, or a T-reg cell disclosed by this document, or an irradiated cell disclosed by this document, or an IFN- ⁇ -pre-stimulated cell disclosed by this document, or an irradiated IFN- ⁇ -pre-stimulated cell disclosed by this document, or an IFN- ⁇ -pre-stimulated irradiated cell disclosed by this document, or a combination thereof), and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S.
  • compositions refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic agent is administered.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic agent is administered.
  • the composition if desired, can also contain minor amounts of pH buffering agents. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E.W. Martin.
  • Such compositions will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject.
  • the formulation should suit the mode of administration.
  • the pharmaceutical compositions are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
  • the pharmaceutical composition of the invention may be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as lyophilized preparations, liquids solutions or suspensions, injectable and infusible solutions, etc.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • the administration of the cell population disclosed by this document, or the pharmaceutical composition comprising same, to the subject in need thereof can be carried out by conventional means.
  • said cell population is administered to the subject by a method which involves transferring the cells to the desired tissue, either in vitro (e.g., as a graft prior to implantation or engrafting) or in vivo , to the animal tissue directly.
  • the cells can be transferred to the desired tissue by any appropriate method, which generally will vary according to the tissue type.
  • cells can be transferred to graft by bathing the graft (or infusing it) with culture medium containing the cells.
  • the cells can be seeded onto the desired site within the tissue to establish a population.
  • Cells can be transferred to sites in vivo using devices such as catheters, trocars, cannulae, stents (which can be seeded with the cells), etc.
  • the cells disclosed by this document can be irradiated before administration to the subject. This treatment makes cells incapable to proliferate or survive for long time periods in the subject.
  • the pharmaceutical composition described herein comprises irradiated cells disclosed by this document.
  • the cells disclosed by this document can be pre-stimulated with IFN- ⁇ , prior to administration to the subject in order to reduce the time period between cell administration and IDO expression in the subject.
  • the pharmaceutical composition described herein comprises IFN- ⁇ -pre-stimulated cells disclosed by this document.
  • the cells disclosed by this document can be both irradiated and pre-stimulated with IFN- ⁇ , in any order, prior to administration to the subject.
  • the pharmaceutical composition described herein comprises irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document or IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document.
  • the cell populations disclosed by this document and pharmaceutical compositions of the invention can be used in a combination therapy.
  • the combination therapy is administered to a subject with an inflammatory disorder that is refractory to one or more anti-inflammatory agents.
  • the combination therapy is used in conjunction with other types of anti-inflammatory agents including, but not limited to, non-steroidal anti-inflammatory drugs (NSAIDs), steroidal anti-inflammatory drugs, beta-agonists, anticholingeric agents, and methyl xanthines.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • beta-agonists beta-agonists
  • anticholingeric agents methyl xanthines
  • NSAIDs include, but are not limited to, ibuprofen, celecoxib, diclofenac, etodolac, fenoprofen, indomethacin, ketoralac, oxaprozin, nabumentone, sulindac, tolmentin, rofecoxib, naproxen, ketoprofen, nabumetone, etc.
  • NSAIDs function by inhibiting a cyclooxgenase enzyme (e.g., COX-1 and/or COX-2).
  • steroidal anti-inflammatory drugs include, but are not limited to, glucocorticoids, dexamethasone, cortisone, hydrocortisone, prednisone, prednisolone, triamcinolone, azulfidine, and eicosanoids such as thromboxanes, and leukotrienes.
  • Monoclonal antibodies, such as Infliximab, can also be used.
  • the combination therapies disclosed by this document can be used prior to, concurrently or subsequent to the administration of such anti-inflammatory agents.
  • anti-inflammatory agents do not encompass agents characterized herein as lymphoid tissue inducers and/or immunomodulatory agents.
  • IDO upon stimulation with IFN- ⁇ can be used for distinguishing cells which express said enzyme from cells which do not express IDO.
  • the document describes a method for distinguishing adult multipotent cells from differentiated cells comprising the step of verifying whether the multipotent cell expresses IDO upon stimulation with IFN- ⁇ .
  • the determination of IDO upon stimulation with IFN- ⁇ can be carried by any conventional technique; in a disclosure, the determination of IDO upon stimulation with IFN- ⁇ can be carried out as disclosed in Example 2.
  • the cells of the cell population disclosed by this document are characterized in that they do not express IDO constitutively, but only upon stimulation with IFN- ⁇ . Moreover, aside from IFN- ⁇ no other pro-inflammatory molecule such us IL-1, TNF- ⁇ , or endotoxin is able to induce by itself the expression of IDO in the cells of the cell population disclosed by this document. This feature can be used for distinguishing the cells of the cell population disclosed by this document from other cells.
  • the document describes a kit comprising a cell population containing (i) cells disclosed by this document and/or (ii) T-reg cells disclosed by this document and/or (iii) irradiated cells disclosed by this document and/or (iv) IFN- ⁇ -pre-stimulated cells disclosed by this document ,and/or (v) irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document, and/or (vi) IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document.
  • Kits disclosed by this document may comprise one, two, three, four, five or all of such cell types.
  • the present document describes the use of a cell population containing cells disclosed by this document, T-reg cells population disclosed by this document, irradiated cells disclosed by this document, IFN- ⁇ -pre-stimulated cells disclosed by this document, irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document, or IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document for preventing, treating, or ameliorating one or more symptoms associated with autoimmune diseases, inflammatory disorders, or immunologically mediated diseases including rejection of transplanted organs and tissues.
  • said cell populations may be used for inducing transplantation tolerance, or for treating, and thereby alleviating, symptoms of autoimmune or inflammatory disorders, or immunologically mediated diseases in a subject suffering from said disorders or diseases.
  • autoimmune diseases and inflammatory diseases have been previously mentioned.
  • disease is an inflammatory disease, such as a chronic inflammatory disease, e.g., IBD or RA.
  • the present document describes methods of preventing, treating, or ameliorating one or more symptoms associated with autoimmune diseases, inflammatory disorders, or immunologically mediated diseases, in a subject suffering from said disorders or diseases, which comprises administering to said subject in need of such treatment of a prophylactically or therapeutically effective amount of a cell population containing cells disclosed by this document, T-reg cells disclosed by this document, irradiated cells disclosed by this document, IFN- ⁇ -pre-stimulated cells disclosed by this document, irradiated IFN- ⁇ -pre-stimulated cells disclosed by this document, or IFN- ⁇ -pre-stimulated irradiated cells disclosed by this document.
  • said cell populations may be used for inducing transplantation tolerance, or for treating, and thereby alleviating, symptoms of autoimmune or inflammatory disorders, or immunologically mediated diseases in a subject suffering from said disorders or diseases.
  • autoimmune diseases and inflammatory diseases have been previously mentioned.
  • disease is an inflammatory disease, such as a chronic inflammatory disease, e.g., IBD or RA.
  • Human adipose tissue was obtained by liposuction, under local anaesthesia and general sedation.
  • a hollow blunt-tipped cannula was introduced into the subcutaneous space through a small incision (less than 0.5 cm in diameter). With gentle suction, the cannula was moved through the adipose tissue abdominal-wall compartment for mechanical disruption of the fatty tissue.
  • a saline solution and the vasoconstrictor epinephrine were injected into the adipose tissue compartment to minimize blood loss. In this way, 80 to 100 ml of raw lipoaspirate were obtained from each patient to be treated.
  • the raw lipoaspirate was washed extensively with sterile phosphate-buffered saline (PBS; Gibco BRL, Paisley, Scotland, UK) to remove blood cells, saline and local anaesthetic.
  • PBS sterile phosphate-buffered saline
  • the extracellular matrix was digested with a solution of type II collagenase (0.075%; Gibco BRL) in balanced salt solution (5 mg/ml; Sigma, St. Louis, USA) for 30 minutes at 37°C to release the cellular fraction. Then the collagenase was inactivated by addition of an equal volume of cell culture medium (Dulbecco's modified Eagle's medium (DMEM; Gibco BRL) that contained 10% fetal bovine serum (FBS; Gibco BRL).
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • the suspension of cells was centrifuged at 250 x g for 10 minutes. Cells were resuspended in 0.16 M NH 4 Cl and allowed to stand for 5 minutes at room temperature (RT) for lysis of erythrocytes. The mixture was centrifuged at 250 x g, and cells were resuspended in DMEM plus 10% FBS and 1% ampicillin/streptomycin mixture (Gibco BRL) and then they were filtered through a 40 ⁇ m mesh and were plated in tissue culture flasks at a concentration of 10-30 x 10 3 cells/cm 2 .
  • Human hyaline articular cartilage was obtained from the knee joint of a donor by means of arthroscopic techniques. About 4 cm 2 of cartilage were taken from the external margin of the phemoral condile, but the size of the biopsy may vary depending on the donor's age, the structure of the articulation and the surgeon's consideration. The biopsy was suspended in a sterile saline solution and stored at 3-8°C until its use. Live cartilage samples should not be stored for more than 48 hours.
  • the cartilage biopsy was transferred to 1 ml of sterile cell culture medium containing 1% FBS, and minced to obtain tissue fragments as small as possible.
  • the resulting cartilage fragments were suspended in a similar medium containing 0.1% (w/v) collagenase, and incubated at 37°C with continuous and gentle agitation. After the digestion, the cell suspension obtained was filtered through a 40 ⁇ m mesh and the cells were plated onto tissue culture flasks at a concentration of 10-30 x 10 3 cells/cm 2 .
  • Cells both from adipose tissue and articular cartilage were separately cultured for 24 hours at 37°C in an atmosphere of 5% CO 2 in air. Then, the culture flasks were washed with PBS to remove non-adhering cells and cell fragments. The cells were maintained in culture in the same medium and under the same conditions until they reached approximately 80% confluence, with replacement of the culture medium every 3 to 4 days. Cells were then passaged with trypsin-EDTA (Gibco BRL) at a dilution of 1:3 which corresponds to a cell density of approximately about 5-6 x 10 3 cells/cm 2 . The cellular growth kinetics of the cells isolated from human adipose tissue and cultured ex vivo for more than 25 cell population doublings is shown in Figure 1 .
  • Cell characterization was performed using cells at culture passages 1 to 25.
  • Cells both from adipose tissue and articular cartilage were analyzed by means of flow cytometry by using antibodies labeled with a fluorescent marker (i.e., by fluorescence immunocytometry) for the presence/absence of a series of surface markers, which included:
  • the antibodies used in the flow cytometry assay were the following:
  • IDO indolamine 2,3-dioxygenase
  • IFN- ⁇ interferon-gamma
  • Example 1 The cells disclosed by this document isolated from human adipose tissue (Example 1), were seeded onto tissue culture plates at a density of 10,000 cells/cm 2 , and incubated for 48 hours in the conditions previously described for cell expansion. Then, different pro-inflammatory stimuli were added to the culture medium, including:
  • the cells were incubated in the presence of the corresponding stimulus for periods ranging from 30 minutes to 48 hours, and then they were collected by trypsin digestion, and lysed in RIPA buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM PMSF (phenyl-methylsulphonylfluoride), 1 mM EDTA (ethylenediaminetetraacetic acid), 5 ⁇ g/ml Aprotinin, 5 ⁇ g/ml Leupeptin, 1% Triton x-100, 1% Sodium deoxycholate, 0.1% SDS) containing protease inhibitors.
  • RIPA buffer 50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM PMSF (phenyl-methylsulphonylfluoride), 1 mM EDTA (ethylenediaminetetraacetic acid), 5 ⁇ g/ml Aprotinin, 5 ⁇ g/ml Leupeptin, 1%
  • mice were cultivated for 2-7 weeks prior to the subcutaneous implantation in immunodeficient mice (5 x 10 6 cells/mouse). The mice were nu / nu strain obtained from Charles River Laboratories. Mice lacked thymus and were T-cell deficient. The implanted mice were followed-up for 4 months prior to sacrifice and pathological study.
  • Pathological study A necropsy was performed on all animals. The animals were examined for gross abnormalities in the brain, lungs, heart, liver, kidneys, spleen, abdominal lymph nodes and injection site. Tissues were collected for a histological examination (parafin section and hematoxilin-eosin (H&E) staining), including injection site, lungs and lymph nodes.
  • H&E hematoxilin-eosin
  • the teratome cellular line (N-TERA) was used as a positive control, which was implanted under identical conditions.
  • Colitis was induced in Balb/c mice (6-8 weeks old, Jackson Laboratories, Bar Harbor, ME) as previously described ( Neurath, M.F., et al. 1995. Antibodies to IL-12 abrogate established experimental colitis in mice. J. Exp. Med. 182, 1281-1290 ). In brief, mice were lightly anesthetized with halothane, and a 3.5 F catheter was inserted intrarectally 4 cm from the anus. To induce colitis, 100 ⁇ l of 50 or 30 mg/ml of TNBS (2,4,6-trinitrobenzene sulfonic acid) (Sigma Chemical Co, St.
  • TNBS 2,4,6-trinitrobenzene sulfonic acid
  • mice received 50% ethanol alone (100 ⁇ l).
  • Animals were treated intrarectally with different numbers of the cells disclosed by this document obtained from human adipose tissue as described in Example 1 (0.3 x 10 6 and 1 x 10 6 cells, suspended in phosphate-buffered saline, PBS) 12 hours after TNBS instillation. In some experiments, said cells were pretreated with 200 U/ml IFN- ⁇ for 24 hours before injection. Animals were monitored daily for survival, appearance of diarrhea, and loss of body weight ( Figures 5 , 6 and 7 ).
  • IFN- ⁇ -pre-stimulated cells showed a faster and stronger recuperation from the TNBS treatment than non-prestimulated cells ( Figure 7 ).
  • the graph shows that TNBS treated mice lost weight dramatically and a clear improvement in mice that received cells.
  • mice-additional experiments Treatment of experimentally-induced inflammatory bowel disease (IBD) in mice-additional experiments: .
  • IBD experimentally-induced inflammatory bowel disease
  • Colitis was induced in Balb/c mice (6-8 weeks old, Jackson Laboratories, Bar Harbor, ME) as previously described ( Neurath, M.F., et al. 1995. Antibodies to IL-12 abrogate established experimental colitis in mice. J. Exp. Med. 182, 1281-1290 ). In brief, mice were lightly anesthetized with halothane, and a 3.5 F catheter was inserted intrarectally 4 cm from the anus. To induce colitis, 100 ⁇ l of 50 or 30 mg/ml of TNBS (2,4,6-trinitrobenzene sulfonic acid) (Sigma Chemical Co, St.
  • TNBS 2,4,6-trinitrobenzene sulfonic acid
  • mice received 50% ethanol alone (100 ⁇ l). Animals were treated intrarectally or intraperitoneally (i.p.) with different numbers of the cells disclosed by this document obtained from human adipose tissue (ASC) as described in Example 1 (0.3 x 10 6 and 1 x 10 6 cells, suspended in phosphate-buffered saline, PBS) 12 hours after TNBS instillation. In some experiments, said cells were pretreated with 200 U/ml IFN- ⁇ for 24 hours before injection.
  • ASC adipose tissue
  • CSFE a fluorescent probe
  • mice were labeled with CFSE (a fluorescent probe) before administration to the mice.
  • Animals were monitored daily for survival, appearance and severity of diarrhea, and loss of body weight.
  • Serum was collected and protein extracts were obtained from colons at the acute phase of the disease (day 3). Cytokine/chemokine contents in protein extracts and in serum were determined by ELISA.
  • the presence of CSFE-labeled cells in the mesenteric lymph nodes were analyzed by flow cytometry.
  • mice treated with the cells disclosed by this document showed a clear improvement in their inflammatory symptoms compared with non-treated animals.
  • the improvement was dose-dependent and statistically significant in all parameters tested, when cells were administered locally (intra-rectally) or systemically (i.p.), although this last route seems to be more effective.
  • Figure 5 there was a dose-dependent improvement of weight gained after the administration of the cells disclosed by the document. Indeed, a dose dependency can be observed in Figures 6 , 7 and 8 with 1x10 6 cells showing a stronger effect than 0.3x10 6 cells. In both cases the cells improved the survival rate of the TNBS treated mice significantly.
  • IFN- ⁇ -pre-stimulated cells showed a faster and stronger recuperation from the TNBS treatment than non-prestimulated cells ( Figure 8 ).
  • the graph shows that TNBS treated mice lost weight dramatically and a clear improvement in mice that received cells. This improvement was also measurable by the severity of colitis.
  • the inflammatory immune response is clearly diminished in animals treated with the cells disclosed by the document.
  • all proinflammatory cytokines (TNF-a, IL-6, IL-1b, IL-12, and IFN ⁇ ) and chemokines (MIP-2 and RANTES) tested, both in the colon (local response) and in the serum (systemic response), were lower in cell-treated animals compared with the non-treated mice.
  • This inhibitory response was enhanced in animals treated with cells pre-stimulated with IFN ⁇ .
  • the immunoregulatory cytokine IL-10 was increased in the colon of ASC-treated mice, comparing with both non-treated TNBS-injured and control animals.
  • neutrophil infiltration as measured by MPO activity was lower in ASC-treated animals, and even lower when cells were pre-stimulated with IFN ⁇ ( Figure 10 ).
  • the labeled cells were localized in the draining lymph nodes of treated animals by means of cell cytometry ( Figure 11 ). This is the localization expected if the administered cells were functioning as APCs.
  • the cells disclosed by this document were obtained from human subcutaneous adipose tissue (ASCs) as described in Example 1. After a minimum of 3 culture passages, the cells were incubated in standard culture medium or in culture medium containing 3 ng/ml IFN ⁇ for 4 days. After that, the cells were stained for some surface markers related with the immune response (specifically related with the activity of antigen presenting cells (APCs)). These markers included the following:
  • the cells disclosed by this document induce the expression of HLA-II, PD-L1 and PD-L2, and a strong upregulation of CD40 and ICAM-1.
  • the results of this experiment are shown in Figure 12 .
  • mice were induced in DBA1/Jlac male mice (6-8 weeks of age) by injecting subcutaneously (s.c.) an emulsion containing 200 ⁇ g of chicken type II collagen (CII) in complete Freund's adjuvant (CFA) and 200 ⁇ g of Mycobacterium tuberculosis H37RA.
  • CII chicken type II collagen
  • CFA complete Freund's adjuvant
  • CIA was followed daily by two different technicians, by measuring the inflammation-redness-ankylosis of the joints of upper and lower limbs, according to a pre-established scoring system.
  • CIA mice were injected intrarticularly (i.a.) once in one of the affected joints.
  • the evolution of the treated animals were followed as previously described, and at day 50 p.i. they were euthanized.
  • Several parameters were measured in the blood and the joints, including: joint cytokines, serum cytokines, immunoglobin isotypes, as well as phenotype and cytokine production of lymphocytes.
  • the cells disclosed by this document clearly decrease CIA incidence and severity in the mouse model.
  • the effect on the immune response is consistent with a strong inhibition of the Th1 response (IFN- ⁇ , TNF ⁇ , IL-2, IL-1 ⁇ , IL-6, IL-12, MIP2, RANTES, and IgG2a) without any increase in the Th2 response (IL-4, IgG1), and with the induction of high levels of immunmoregulatory cytokines (IL-10 and TGF- ⁇ ).
  • effector CD25 - Foxp3 -
  • regulatory T cells CD4 + CD25 + Foxp3 +
  • a proliferative assay was performed in which autorreactive T cells isolated from CIA mice were co-cultured with increasing numbers of DLN T cells (regulatory T cells) from untreated (control) or ASC-treated CIA mice (ratios from 1/64 to 1/1), and stimulated with CII (10 ⁇ g/ml) and splenic APCs.
  • both the DLN and the synovial membrane of CIA mice treated with the cells disclosed by this document induce an increase in the numbers of regulatory T cells (CD4 + CD25 + Foxp3 + ), without any increase in the numbers of effector T cells, comparing with the untreated (control) CIA mice.
  • Adipose derived ASCs disclosed by the document, obtained via the methods of Example 1, were plated at 5000 cells/cm 2 with and without 200.000 lymphocytes (activated w/ 10 ⁇ gPHA/ml) and co-cultured for 3 days. Proliferation of the lymphocytes was measured by H3 incorporation. As shown in Figure 17 , the co-culture of ASCs and lymphocytes resulted in an 86% inhibition of lymphocytes proliferation. The addition different concentrations of 1Methyl-trytophan (1-MT) reverted this suppression. 1-MT is a non-metabolizable tryptophan analog. The assay demonstrates the necessity of tryptophan catabolism via IDO to induce the immuno-suppressive activity of the cells disclosed by the document.
  • Fresh solutions in the range of 100 ⁇ M to 100mM of trytophan and kynurenine were prepared in 10% acetonitrile in potassium-phosphate buffer (50mM pH 6.0). From these stock solutions 50 ⁇ l tryptophan and 10 ⁇ l kynurenine and 940 ⁇ l BSA (70g/l) or 10%FCS were combined to make up the control sample and stored at -80°C Sample preparation: 200 ⁇ l or more of supernatant from samples (cell cultures) were collected in Eppendorf tubes and stored at -80°C. Samples and control samples were thawed and 200 ⁇ l 50mM potassium-phosphate buffer pH6.0 was added to each 200 ⁇ l sample in an Eppendorf tube.
  • the HPLC column was prepared as known in the art and equilibrated with mobile phase, which consisted of 40mM sodium-citrate pH5 - in 5% acteonitrile 50 ⁇ l of above described sample of 150 ⁇ l sample was injected into the column (C18 reverse phase). Separation occurs by an isocratic flow rate of 700 ⁇ l/min.
  • the photometric detection of L-kynurenine occurs at 365nm, for L-tryptophan at 280nm.
  • ASCs plated at 5000cell/cm 2 and stimulated at 3ng/ml IFN- ⁇ for up to 120 hours produce IDO, the activity of which is measured by the metabolisation of tryptophan and production of kynurenine using HPLC.
  • ASCs plated at 5000cell/cm 2 and stimulated at 3pg/ml IFN- ⁇ for up to 120hours did not produce IDO. No kynurenine could be detected ( Figure 19 ).
  • ASCs plated at 500cell/cm 2 and stimulated at 3ng/ml IFN- ⁇ for up to 120 hours did not produce significant amounts of IDO ( Figure 20 ).
  • 4kda- Dextran-FITC (Sigma) was added to the cells of Example 1 for 24 hours in culture. The cell were washed and analyzed for the incorporation of the fluorescent FITC.
  • Figure 21A shows the cells bright field image of the washed cell population.
  • Figure 21B shows the same population using fluorescence microscopy using Green Fluorescent Protein filters known in the art.
  • the uptake of the fluorescent marker visible in Figure 21 B shows that the cells are able to phagocytose small weight molecules and this indicates that these cells are capable of antigen-presentation via HLA class II induced by additional treatment of the cells with IFN- ⁇ .

Claims (8)

  1. Population isolierter Zellen aus Fettgewebe, dadurch gekennzeichnet, dass die Zellen der Zellpopulation
    i) für Antigen-präsentierende Zellen (APC) spezifische Marker nicht exprimieren, wobei die für APC spezifische Marker CD11b, CD11c, CD14, CD45 und HLAII sind,
    ii) Indolamin-2,3-Dioxygenase (IDO) nicht konstitutiv exprimieren,
    iii) IDO aufgrund Stimulierung mit Interferon-y (IFN-γ) exprimieren,
    und
    iv) die Fähigkeit aufweisen, in mindestens zwei Zelllinien zu differenzieren,
    zur Verwendung bei der Vorbeugung, Behandlung oder Verbesserung eines Symptoms oder mehrerer Symptome, die mit Chronisch-entzündlicher Darmerkrankung (IBD) in einem Patient, der an IDB leidet, assoziiert sind.
  2. Eine pharmazeutische Zusammensetzung enthaltend eine Population isolierter Zellen aus Fettgewebe, dadurch gekennzeichnet, dass die Zellen der Zellpopulation
    i) für Antigen-präsentierende Zellen (APC) spezifische Marker nicht exprimieren, wobei die für APC spezifische Marker CD11b, CD11c, CD14, CD45 oder HLAII sind,
    ii) Indolamin-2,3-Dioxygenase (IDO) nicht konstitutiv exprimieren,
    iii) IDO aufgrund Stimulierung mit Interferon-y (IFN-γ) exprimieren,
    und
    iv) die Fähigkeit aufweisen, in mindestens zwei Zelllinien zu differenzieren,
    und einen pharmazeutisch akzeptablen Träger zur Verwendung bei der Vorbeugung, Behandlung oder Verbesserung eines Symptoms oder mehrerer Symptome, die mit Chronisch-entzündlicher Darmerkrankung (IBD) in einem Patient, der an IDB leidet, assoziiert sind.
  3. Einen Kit enthaltend eine Population isolierter Zellen aus Fettgewebe, dadurch gekennzeichnet, dass die Zellen der Zellpopulation
    i) für Antigen-präsentierende Zellen (APC) spezifische Marker nicht exprimieren, wobei die für APC spezifische Marker CD11b, CD11c, CD14, CD45 oder HLAII sind,
    ii) Indolamin-2,3-Dioxygenase (IDO) nicht konstitutiv exprimieren,
    iii) IDO aufgrund Stimulierung mit Interferon-y (IFN-γ) exprimieren,
    und
    iv) die Fähigkeit aufweisen, in mindestens zwei Zelllinien zu differenzieren,
    zur Verwendung bei der Vorbeugung, Behandlung oder Verbesserung eines Symptoms oder mehrerer Symptome, die mit Chronisch-entzündlicher Darmerkrankung (IBD) in einem Patient, der an IDB leidet, assoziiert sind.
  4. Verwendung einer Population isolierter Zellen aus Fettgewebe, dadurch gekennzeichnet, dass die Zellen der Zellpopulation
    i) für Antigen-präsentierende Zellen (APC) spezifische Marker nicht exprimieren, wobei die für APC spezifische Marker CD11b, CD11c, CD14, CD45 oder HLAII sind,
    ii) Indolamin-2,3-Dioxygenase (IDO) nicht konstitutiv exprimieren,
    iii) IDO aufgrund Stimulierung mit Interferon-y (IFN-γ) exprimieren,
    und
    iv) die Fähigkeit aufweisen, in mindestens zwei Zelllinien zu differenzieren,
    zur Herstellung eines Arzneimittels zur Verwendung bei der Vorbeugung, Behandlung oder Verbesserung eines Symptoms oder mehrerer Symptome, die mit Chronisch-entzündlicher Darmerkrankung (IBD) in einem Patient, der an IDB leidet, assoziiert sind.
  5. Population isolierter Zellen zur Verwendung gemäß den Ansprüchen 1, pharmazeutische Zusammensetzung zur Verwendung gemäß Anspruch 2, Kit zur Verwendung gemäß Anspruch 4 oder Verwendung gemäß Anspruch 4, dadurch gekennzeichnet, dass die Zellpopulation negativ ist bezüglich der folgenden Zelloberflächenmarker: CD11b, CD11c, CD14, CD31, CD34, CD45, CD133 und HLAII.
  6. Population isolierter Zellen zur Verwendung gemäß den Ansprüchen 1, pharmazeutische Zusammensetzung zur Verwendung gemäß Anspruch 2, Kit zur Verwendung gemäß Anspruch 4 oder Verwendung gemäß Anspruch 4, dadurch gekennzeichnet, dass die Zellpopulation positiv ist für mindestens einen und vorzugsweise für alle der folgenden Zelloberflächenmarker: CD9, CD44, CD54, CD90 und CD105.
  7. Population isolierter Zellen zur Verwendung gemäß den Ansprüchen 1, pharmazeutische Zusammensetzung zur Verwendung gemäß Anspruch 2, Kit zur Verwendung gemäß Anspruch 4 oder Verwendung gemäß Anspruch 4, dadurch gekennzeichnet, dass die Zellpopulation fähig ist, ex vivo vermehrt zu werden.
  8. Population isolierter Zellen zur Verwendung gemäß den Ansprüchen 1, pharmazeutische Zusammensetzung zur Verwendung gemäß Anspruch 2, Kit zur Verwendung gemäß Anspruch 4 oder Verwendung gemäß Anspruch 4, dadurch gekennzeichnet, dass die Zellpopulation mindestens ein antigenes Polypeptid exprimiert.
EP06777197.2A 2005-09-23 2006-09-22 Zellpopulationen mit immunregulationsaktivität, verfahren zur isolierung und verwendungen Active EP1926813B2 (de)

Priority Applications (5)

Application Number Priority Date Filing Date Title
SI200632094T SI1926813T2 (sl) 2005-09-23 2006-09-22 Celična populacija z imunoregulatorno aktivnostjo, postopek za izolacijo in uporabe
EP10195268.7A EP2340847A3 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunoregulierenden Aktivität, Verfahren zur Isolierung und Verwendung
EP17154940.5A EP3184631B1 (de) 2005-09-23 2006-09-22 Zellenpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen
PL06777197T PL1926813T5 (pl) 2005-09-23 2006-09-22 Populacje komórkowe mające aktywność immunoregulatorową, sposób izolacji i zastosowania
EP17154939.7A EP3184630B1 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP05077186 2005-09-23
PCT/EP2006/009244 WO2007039150A2 (en) 2005-09-23 2006-09-22 Cell populations having immunoregulatory activity, method for isolation and uses

Related Child Applications (6)

Application Number Title Priority Date Filing Date
EP10195268.7A Division-Into EP2340847A3 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunoregulierenden Aktivität, Verfahren zur Isolierung und Verwendung
EP10195268.7A Division EP2340847A3 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunoregulierenden Aktivität, Verfahren zur Isolierung und Verwendung
EP17154940.5A Division-Into EP3184631B1 (de) 2005-09-23 2006-09-22 Zellenpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen
EP17154940.5A Division EP3184631B1 (de) 2005-09-23 2006-09-22 Zellenpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen
EP17154939.7A Division-Into EP3184630B1 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen
EP17154939.7A Division EP3184630B1 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen

Publications (3)

Publication Number Publication Date
EP1926813A2 EP1926813A2 (de) 2008-06-04
EP1926813B1 EP1926813B1 (de) 2016-06-08
EP1926813B2 true EP1926813B2 (de) 2019-05-22

Family

ID=37708240

Family Applications (4)

Application Number Title Priority Date Filing Date
EP06777197.2A Active EP1926813B2 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunregulationsaktivität, verfahren zur isolierung und verwendungen
EP10195268.7A Pending EP2340847A3 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunoregulierenden Aktivität, Verfahren zur Isolierung und Verwendung
EP17154939.7A Active EP3184630B1 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen
EP17154940.5A Active EP3184631B1 (de) 2005-09-23 2006-09-22 Zellenpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen

Family Applications After (3)

Application Number Title Priority Date Filing Date
EP10195268.7A Pending EP2340847A3 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunoregulierenden Aktivität, Verfahren zur Isolierung und Verwendung
EP17154939.7A Active EP3184630B1 (de) 2005-09-23 2006-09-22 Zellpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen
EP17154940.5A Active EP3184631B1 (de) 2005-09-23 2006-09-22 Zellenpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen

Country Status (19)

Country Link
US (5) US20090130067A1 (de)
EP (4) EP1926813B2 (de)
JP (2) JP5925408B2 (de)
KR (2) KR101536239B1 (de)
CN (2) CN107723274B (de)
AU (1) AU2006299144A1 (de)
CA (2) CA2953782C (de)
CY (1) CY1117972T1 (de)
DK (1) DK1926813T4 (de)
ES (2) ES2751692T3 (de)
HU (1) HUE030503T2 (de)
IL (3) IL190363A0 (de)
LT (1) LT1926813T (de)
MX (1) MX342474B (de)
PL (1) PL1926813T5 (de)
PT (1) PT1926813T (de)
SG (3) SG10201400793RA (de)
SI (1) SI1926813T2 (de)
WO (1) WO2007039150A2 (de)

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060045872A1 (en) 2004-08-25 2006-03-02 Universidad Autonoma De Madrid Ciudad Universitaria de Cantoblanco Use of adipose tissue-derived stromal stem cells in treating fistula
WO2007039150A2 (en) 2005-09-23 2007-04-12 Cellerix, S.L. Cell populations having immunoregulatory activity, method for isolation and uses
PL2347775T3 (pl) 2005-12-13 2020-11-16 President And Fellows Of Harvard College Rusztowania do przeszczepiania komórek
US7989173B2 (en) 2006-12-27 2011-08-02 The Johns Hopkins University Detection and diagnosis of inflammatory disorders
JP5690143B2 (ja) 2008-02-13 2015-03-25 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ 持続的細胞プログラミング装置
CN102036671A (zh) 2008-03-28 2011-04-27 永生细胞生技股份有限公司 使用脐带血细胞治疗脑损伤
CA2721150C (en) * 2008-04-17 2017-09-26 Herlev Hospital Indoleamine 2,3-dioxygenase based immunotherapy
GB0814249D0 (en) * 2008-08-04 2008-09-10 Cellerix Sa Uses of mesenchymal stem cells
AU2009201915C1 (en) 2008-08-22 2015-02-26 Regeneus Ltd Therapeutic methods
EP2204442A1 (de) * 2008-10-01 2010-07-07 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Verfahren zur Anreicherung von Stammzellen in einer Kultur
GB0820397D0 (en) 2008-11-07 2008-12-17 Cellerix Sa Cells, nucleic acid constructs, cells comprising said constructs and methods utilizing said cells in the treatment of diseases
WO2011004264A1 (en) 2009-07-09 2011-01-13 Cellerix Sa Methods and compositions for use in cellular therapies
AU2010278702C1 (en) * 2009-07-31 2016-07-14 Forsyth Dental Infirmary For Children Programming of cells for tolerogenic therapies
IN2012DN02753A (de) 2009-08-31 2015-09-18 Amplimmune Inc
GB0918615D0 (en) 2009-10-23 2009-12-09 Cellerix Sa Cell populations having immunoregulatory activity, methods for the preparation and uses thereof
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
CA2813751C (en) 2010-10-06 2019-11-12 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
WO2012095743A2 (en) 2011-01-12 2012-07-19 Cellerix Sa Methods and compositions for use in intralymphatic cellular therapies
JP2014508527A (ja) * 2011-03-11 2014-04-10 ティゲニクス、エセ、ア、ウ 免疫調節活性を有する細胞集団、単離方法および使用
ES2661454T3 (es) * 2011-04-25 2018-04-02 University Of Southern California Composiciones para la regeneración mejorada de tejidos por supresión de interferón-gamma y factor de necrosis tumoral-alfa
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
WO2012156522A1 (en) 2011-05-19 2012-11-22 Tigenix Sa Cell populations having immunoregulatory activity, methods for the preparation and uses thereof
CA2870309C (en) 2012-04-16 2024-02-20 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
JP6412933B2 (ja) * 2013-06-25 2018-10-24 ティジェニクス エス.エー.ユー. 免疫調節活性を有する細胞集団、その調製方法および使用
EP3027235A1 (de) 2013-07-30 2016-06-08 Musculoskeletal Transplant Foundation Aus einem azellulären weichgewebe gewonnene matrizen und verfahren zur herstellung davon
AU2014348683B2 (en) 2013-11-18 2020-11-05 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
AU2015241422B2 (en) 2014-04-01 2020-12-03 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
WO2015168379A2 (en) 2014-04-30 2015-11-05 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
BR112016030758B8 (pt) 2014-06-30 2022-10-25 Tigenix S A U Uso de células estromais multipotentes derivadas de tecido adiposo mesenquimal
EP3160480B1 (de) 2014-06-30 2020-09-09 TiGenix, S.A.U. Mesenchymale stromazellen zur behandlung von rheumatoider arthritis
JP2016017068A (ja) * 2014-07-10 2016-02-01 国立大学法人金沢大学 脂肪組織由来間質細胞群を含む消化管炎症治療剤
KR101675521B1 (ko) * 2014-07-11 2016-11-17 대한민국 인돌아민 2,3-디옥시게나아제를 이용하여 인터페론-감마의 발현을 조절하는 방법
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
CN107708756A (zh) 2015-04-10 2018-02-16 哈佛学院院长等 免疫细胞捕获装置及其制备和使用方法
WO2016187413A1 (en) 2015-05-21 2016-11-24 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
EP3411475A4 (de) 2016-02-06 2019-09-11 President and Fellows of Harvard College Rekapitulation der hämatopoietischen nische zur wiederherstellung der immunität
WO2017144552A1 (en) 2016-02-22 2017-08-31 Centauri Biotech. S.L. Pharmaceutical or veterinary cell compositions comprising mesenchymal stromal cells (mscs) and dimethyl sulfoxide (dmso)
GB201604304D0 (en) 2016-03-14 2016-04-27 Tigenix S A U Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in crohn's disease
CN115305229A (zh) 2016-07-13 2022-11-08 哈佛学院院长等 抗原呈递细胞模拟支架及其制备和使用方法
US10300090B2 (en) 2017-03-15 2019-05-28 Orca Biosystems, Inc. Compositions of hematopoietic stem cell transplants
US20210008120A1 (en) 2018-03-13 2021-01-14 Oxacell Ag Cell products with improved stability and uses thereof
US11413314B2 (en) 2018-06-15 2022-08-16 NextPhase Therapeutics, Inc. Pharmaceutical composition for dermatology and uses thereof
CN113490503A (zh) * 2019-05-21 2021-10-08 艾默生物医学股份有限公司 间质干细胞于治疗免疫相关疾病的用途
KR102438419B1 (ko) * 2021-12-01 2022-09-01 메디포스트(주) Cd47 과발현 중간엽 줄기세포를 포함하는 폐 질환 예방 또는 치료용 조성물

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5145770A (en) 1990-06-04 1992-09-08 Biosurface Technology, Inc. Cryopreservation of cultured epithelial sheets
US5733542A (en) * 1990-11-16 1998-03-31 Haynesworth; Stephen E. Enhancing bone marrow engraftment using MSCS
US5709854A (en) 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
ATE247933T1 (de) 1994-06-06 2003-09-15 Univ Case Western Reserve Biomatrix für geweberegenaration
US6174333B1 (en) 1994-06-06 2001-01-16 Osiris Therapeutics, Inc. Biomatrix for soft tissue regeneration using mesenchymal stem cells
US6391297B1 (en) * 1997-12-02 2002-05-21 Artecel Sciences, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
US6395876B1 (en) 1997-12-05 2002-05-28 Medical College Of Georgia Research Institute, Inc. High-affinity tryptophan transporter
US6368636B1 (en) * 1998-03-18 2002-04-09 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
JP2002510655A (ja) * 1998-04-03 2002-04-09 オシリス セラピューティクス,インコーポレイテッド 免疫抑制剤としての間葉幹細胞によるt細胞応答を阻害する方法とその利用
KR100968164B1 (ko) 1999-03-10 2010-07-06 더 리전츠 오브 더 유니버시티 오브 캘리포니아 지방 유래 간세포 및 격자
US6777231B1 (en) 1999-03-10 2004-08-17 The Regents Of The University Of California Adipose-derived stem cells and lattices
US20030082152A1 (en) 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
US20050153442A1 (en) 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
US7670628B2 (en) 1999-07-07 2010-03-02 Angioblast Systems, Inc. Mesenchymal precursor cell
US7078232B2 (en) 1999-08-19 2006-07-18 Artecel, Inc. Adipose tissue-derived adult stem or stromal cells for the repair of articular cartilage fractures and uses thereof
WO2002067867A2 (en) 2001-02-23 2002-09-06 The University Of Pittsburgh Rapid preparation of stem cell matrices for use in tissue and organ treatment and repair
EP1241249A1 (de) * 2001-03-12 2002-09-18 Gerold Schuler Regulatorische CD4+CD25+ T Zellen aus menschlichem Blut
US20030054331A1 (en) 2001-09-14 2003-03-20 Stemsource, Inc. Preservation of non embryonic cells from non hematopoietic tissues
JP2005508393A (ja) * 2001-11-09 2005-03-31 アーテセル・サイエンシズ・インコーポレーテツド 胚および成人幹細胞をサポートするための間質細胞の使用の方法および組成物
WO2003051388A2 (en) * 2001-12-18 2003-06-26 Mondobiotech Laboratories Anstalt Pharmaceutical composition of interferon gamma or pirfenidone with molecular diagnostics for the improved treatment of interstitial lung diseases
US20040204458A1 (en) * 2002-08-16 2004-10-14 Boehringer Ingelheim Pharma Gmbh & Co. Kg Use of Lck inhibitors for treatment of immunologic diseases
WO2004090095A2 (en) * 2003-04-01 2004-10-21 University Of Southern California Generation of human regulatory t cells by bacterial toxins for the treatment of inflammatory disorders
ES2265199B1 (es) * 2003-06-12 2008-02-01 Cellerix, S.L. Celulas madre adultas multipotentes procedentes de condrocitos desdiferenciados y sus aplicaciones.
US9074190B2 (en) 2003-10-07 2015-07-07 Biomaster, Inc. Cell differentiation of adipose-derived precursor cells
ATE493492T1 (de) 2003-11-04 2011-01-15 Biomaster Inc Verfahren und system zur herstellung von stammzellen aus fettgewebe
EP1699523A4 (de) 2003-12-24 2008-05-21 Univ Columbia Erzeugung eines biologischen atrioventrikulären bypass zur kompensierung eines atrioventrikulären blocks
AU2005206746B2 (en) * 2004-01-08 2008-08-28 Regents Of The University Of California Regulatory t cells suppress autoimmunity
WO2005093044A1 (en) * 2004-03-22 2005-10-06 Osiris Therapeutics, Inc. Mesenchymal stem cells and uses therefor
RU2252252C1 (ru) 2004-04-09 2005-05-20 Тепляшин Александр Сергеевич Способ выделения мезенхимальных стволовых клеток
US20060045872A1 (en) 2004-08-25 2006-03-02 Universidad Autonoma De Madrid Ciudad Universitaria de Cantoblanco Use of adipose tissue-derived stromal stem cells in treating fistula
ES2264862B8 (es) 2004-08-25 2017-01-20 Cellerix, S.L. Biomaterial para sutura.
ES2313805B1 (es) 2004-10-04 2009-12-23 Cellerix, S.L. Identificacion y aislamiento de celulas multipotentes de tejido mesenquimal no osteocondral.
WO2007039150A2 (en) 2005-09-23 2007-04-12 Cellerix, S.L. Cell populations having immunoregulatory activity, method for isolation and uses
WO2011004264A1 (en) 2009-07-09 2011-01-13 Cellerix Sa Methods and compositions for use in cellular therapies

Also Published As

Publication number Publication date
KR20080048555A (ko) 2008-06-02
EP1926813B1 (de) 2016-06-08
IL190363A0 (en) 2011-08-01
JP2009508507A (ja) 2009-03-05
CN107723274A (zh) 2018-02-23
SI1926813T1 (sl) 2016-10-28
SG10202001007TA (en) 2020-03-30
CA2953782A1 (en) 2007-04-12
US20150224146A1 (en) 2015-08-13
EP2340847A3 (de) 2016-11-09
DK1926813T3 (en) 2016-09-19
US20170296593A1 (en) 2017-10-19
IL228670A0 (en) 2013-12-31
MX342474B (es) 2016-09-30
EP2340847A2 (de) 2011-07-06
EP3184631B1 (de) 2020-08-12
US20170296592A1 (en) 2017-10-19
EP3184630B1 (de) 2019-07-17
EP1926813A2 (de) 2008-06-04
CA2953782C (en) 2023-09-12
PT1926813T (pt) 2016-09-06
EP3184630A1 (de) 2017-06-28
US9943550B2 (en) 2018-04-17
ES2589311T3 (es) 2016-11-11
ES2751692T3 (es) 2020-04-01
ES2589311T5 (es) 2020-02-14
SG10201400793RA (en) 2014-08-28
US20090130067A1 (en) 2009-05-21
CA2623353C (en) 2017-02-21
IL246494A0 (en) 2016-08-31
JP2014221045A (ja) 2014-11-27
SG165418A1 (en) 2010-10-28
PL1926813T3 (pl) 2017-01-31
SI1926813T2 (sl) 2019-11-29
CN107723274B (zh) 2021-05-07
IL246494B (en) 2021-09-30
WO2007039150A3 (en) 2007-08-23
LT1926813T (lt) 2016-11-10
JP6089004B2 (ja) 2017-03-01
CN101313062A (zh) 2008-11-26
HUE030503T2 (en) 2017-05-29
PL1926813T5 (pl) 2019-11-29
EP3184631A1 (de) 2017-06-28
AU2006299144A1 (en) 2007-04-12
DK1926813T4 (da) 2019-08-19
WO2007039150A2 (en) 2007-04-12
CY1117972T1 (el) 2017-05-17
US20220313742A1 (en) 2022-10-06
JP5925408B2 (ja) 2016-05-25
IL228670A (en) 2017-06-29
KR20140003631A (ko) 2014-01-09
KR101536239B1 (ko) 2015-07-13
CA2623353A1 (en) 2007-04-12

Similar Documents

Publication Publication Date Title
US20220313742A1 (en) Cell populations having immunoregulatory activity, method for isolation and uses
EP2451943B1 (de) Verfahren und zusammensetzungen für zelltherapien
EP2663316A2 (de) Aus fettgewebe stammende mesenchymale stammzellen für die intralymphatische verabreichung bei autoimmun- und entzündungserkrankungen
EP2491115B1 (de) Zellpopulationen mit immunregulatorischer aktivität, verfahren zu ihrer herstellung und verwendung
EP2683813A1 (de) Zellenpopulationen mit immunregulatorischer aktivität, isolierverfahren und verwendungen
AU2018253517B2 (en) Cell populations having immunoregulatory activity, method for isolation and uses
AU2012268272B2 (en) Cell populations having immunoregulatory activity, method for isolation and uses
MX2008003881A (en) Cell populations having immunoregulatory activity, method for isolation and uses

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080331

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

17Q First examination report despatched

Effective date: 20081205

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CELLERIX, S.A.

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602006049302

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C12N0005080000

Ipc: C12N0005078300

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 5/0783 20100101AFI20150930BHEP

Ipc: C12N 5/0775 20100101ALI20150930BHEP

INTG Intention to grant announced

Effective date: 20151023

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS

Owner name: TIGENIX, S.A.U.

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 805267

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160715

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602006049302

Country of ref document: DE

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 1926813

Country of ref document: PT

Date of ref document: 20160906

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20160831

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20160913

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2589311

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20161111

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E012511

Country of ref document: EE

Effective date: 20160927

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20160402148

Country of ref document: GR

Effective date: 20161118

REG Reference to a national code

Ref country code: DE

Ref legal event code: R026

Ref document number: 602006049302

Country of ref document: DE

PLBI Opposition filed

Free format text: ORIGINAL CODE: 0009260

26 Opposition filed

Opponent name: MUELLER, CHRISTIAN

Effective date: 20170308

PLAX Notice of opposition and request to file observation + time limit sent

Free format text: ORIGINAL CODE: EPIDOSNOBS2

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E030503

Country of ref document: HU

PLBB Reply of patent proprietor to notice(s) of opposition received

Free format text: ORIGINAL CODE: EPIDOSNOBS3

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

PLBP Opposition withdrawn

Free format text: ORIGINAL CODE: 0009264

PLAZ Examination of admissibility of opposition: despatch of communication + time limit

Free format text: ORIGINAL CODE: EPIDOSNOPE2

PLAY Examination report in opposition despatched + time limit

Free format text: ORIGINAL CODE: EPIDOSNORE2

PLBC Reply to examination report in opposition received

Free format text: ORIGINAL CODE: EPIDOSNORE3

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 13

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 805267

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160608

PUAH Patent maintained in amended form

Free format text: ORIGINAL CODE: 0009272

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: PATENT MAINTAINED AS AMENDED

REG Reference to a national code

Ref country code: CH

Ref legal event code: AELC

27A Patent maintained in amended form

Effective date: 20190522

AK Designated contracting states

Kind code of ref document: B2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: DE

Ref legal event code: R102

Ref document number: 602006049302

Country of ref document: DE

REG Reference to a national code

Ref country code: BE

Ref legal event code: FP

Effective date: 20160907

REG Reference to a national code

Ref country code: DK

Ref legal event code: T4

Effective date: 20190816

REG Reference to a national code

Ref country code: SE

Ref legal event code: RPEO

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: EE

Ref legal event code: LD4A

Ref document number: E012511

Country of ref document: EE

REG Reference to a national code

Ref country code: SK

Ref legal event code: T5

Ref document number: E 21971

Country of ref document: SK

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20190402532

Country of ref document: GR

Effective date: 20191128

REG Reference to a national code

Ref country code: ES

Ref legal event code: DC2A

Ref document number: 2589311

Country of ref document: ES

Kind code of ref document: T5

Effective date: 20200214

REG Reference to a national code

Ref country code: EE

Ref legal event code: GB1A

Ref document number: E012511

Country of ref document: EE

REG Reference to a national code

Ref country code: LU

Ref legal event code: PD

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS; ES

Free format text: FORMER OWNER: TIGENIX, S.A.U.

Effective date: 20220714

REG Reference to a national code

Ref country code: GB

Ref legal event code: 732E

Free format text: REGISTERED BETWEEN 20220818 AND 20220824

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 602006049302

Country of ref document: DE

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED, OSAKA-S, JP

Free format text: FORMER OWNERS: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS, MADRID, ES; TIGENIX, S.A.U., TRES CANTOS, MADRID, ES

Ref country code: DE

Ref legal event code: R081

Ref document number: 602006049302

Country of ref document: DE

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICA, ES

Free format text: FORMER OWNERS: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS, MADRID, ES; TIGENIX, S.A.U., TRES CANTOS, MADRID, ES

REG Reference to a national code

Ref country code: NL

Ref legal event code: PD

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED; JP

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), ASSIGNMENT; FORMER OWNER NAME: TIGENIX, S.A.U.

Effective date: 20221018

REG Reference to a national code

Ref country code: HU

Ref legal event code: GB9C

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED, JP

Free format text: FORMER OWNER(S): TIGENIX, S.A.U., ES

Ref country code: HU

Ref legal event code: GB9C

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS, ES

Free format text: FORMER OWNER(S): TIGENIX, S.A.U., ES

Ref country code: HU

Ref legal event code: FH1C

Free format text: FORMER REPRESENTATIVE(S): DANUBIA SZABADALMI ES JOGI IRODA KFT., HU

Representative=s name: DANUBIA SZABADALMI ES JOGI IRODA KFT., HU

REG Reference to a national code

Ref country code: SI

Ref legal event code: SP73

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED; ES

Effective date: 20221123

REG Reference to a national code

Ref country code: SK

Ref legal event code: PC4A

Ref document number: E 21971

Country of ref document: SK

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICA, ES

Free format text: FORMER OWNER: TIGENIX, S.A.U., TRES CANTOS (MADRID), ES; CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS, MADRID, ES

Effective date: 20221213

Ref country code: SK

Ref legal event code: PC4A

Ref document number: E 21971

Country of ref document: SK

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED, CHUO-KU, JP

Free format text: FORMER OWNER: TIGENIX, S.A.U., TRES CANTOS (MADRID), ES; CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS, MADRID, ES

Effective date: 20221213

REG Reference to a national code

Ref country code: BE

Ref legal event code: PD

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS; ES

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), ASSIGNMENT; FORMER OWNER NAME: TIGENIX, S.A.U.

Effective date: 20220921

REG Reference to a national code

Ref country code: AT

Ref legal event code: PC

Ref document number: 805267

Country of ref document: AT

Kind code of ref document: T

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICA, ES

Effective date: 20230109

Ref country code: AT

Ref legal event code: PC

Ref document number: 805267

Country of ref document: AT

Kind code of ref document: T

Owner name: TAKEDA PHARMACEUTIKAL COMPANY LIMITED, JP

Effective date: 20230109

REG Reference to a national code

Ref country code: ES

Ref legal event code: PC2A

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED

Effective date: 20230315

REG Reference to a national code

Ref country code: BE

Ref legal event code: PD

Owner name: CONSEJO SUPERIOR DE INVESTIGACIONES CIENTIFICAS; ES

Free format text: DETAILS ASSIGNMENT: CHANGE OF OWNER(S), ASSIGNMENT; FORMER OWNER NAME: TIGENIX, S.A.U.

Effective date: 20220921

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230509

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20230822

Year of fee payment: 18

Ref country code: LU

Payment date: 20230822

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230825

Year of fee payment: 18

Ref country code: RO

Payment date: 20230906

Year of fee payment: 18

Ref country code: MC

Payment date: 20230824

Year of fee payment: 18

Ref country code: IT

Payment date: 20230822

Year of fee payment: 18

Ref country code: IE

Payment date: 20230823

Year of fee payment: 18

Ref country code: GB

Payment date: 20230823

Year of fee payment: 18

Ref country code: FI

Payment date: 20230823

Year of fee payment: 18

Ref country code: EE

Payment date: 20230822

Year of fee payment: 18

Ref country code: CZ

Payment date: 20230825

Year of fee payment: 18

Ref country code: BG

Payment date: 20230828

Year of fee payment: 18

Ref country code: AT

Payment date: 20230823

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SK

Payment date: 20230830

Year of fee payment: 18

Ref country code: SI

Payment date: 20230908

Year of fee payment: 18

Ref country code: SE

Payment date: 20230822

Year of fee payment: 18

Ref country code: PT

Payment date: 20230823

Year of fee payment: 18

Ref country code: PL

Payment date: 20230823

Year of fee payment: 18

Ref country code: IS

Payment date: 20230915

Year of fee payment: 18

Ref country code: HU

Payment date: 20230829

Year of fee payment: 18

Ref country code: GR

Payment date: 20230823

Year of fee payment: 18

Ref country code: FR

Payment date: 20230822

Year of fee payment: 18

Ref country code: DK

Payment date: 20230822

Year of fee payment: 18

Ref country code: DE

Payment date: 20230822

Year of fee payment: 18

Ref country code: BE

Payment date: 20230822

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LV

Payment date: 20230822

Year of fee payment: 18

Ref country code: LT

Payment date: 20230822

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20231002

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CY

Payment date: 20230823

Year of fee payment: 18

Ref country code: CH

Payment date: 20231001

Year of fee payment: 18

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 805267

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190522